1
|
Du X, Zou R, Du K, Huang D, Miao C, Qiu B, Ding W, Li C. Modeling Colorectal Cancer-Induced Liver Portal Vein Microthrombus on a Hepatic Lobule Chip. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 38033197 DOI: 10.1021/acsami.3c14417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Colorectal cancer is one of the most common malignant tumors. At the advanced stage of colorectal cancer, cancer cells migrate with the blood to the liver from the hepatic portal vein, eventually resulting in a portal vein tumor thrombus (PVTT). To date, the progression of the early onset of PVTT [portal vein microthrombus (PVmT) induced by tumors] is unclear. Herein, we developed an on-chip PVmT model by loading the spheroid of colorectal cancer cells into the portal vein of a hepatic lobule chip (HLC). On the HLC, the progression of PVmT was presented, and early changes in metabolites of hepatic cells and in structures of hepatic plates and sinusoids induced by PVmT were analyzed. We replicated intrahepatic angiogenesis, thickened blood vessels, an increased number of hepatocytes, disordered hepatic plates, and decreased concentrations of biomarkers of hepatic cell functions in PVmT progression on a microfluidic chip for the first time. In addition, the combined therapy of thermo-ablation and chemo-drug for PVmT was preliminarily demonstrated. This study provides a promising method for understanding PVTT evolution and offers a valuable reference for PVTT therapy.
Collapse
Affiliation(s)
- Xiaofang Du
- School of Information Science and Technology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Rong Zou
- School of Information Science and Technology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Kun Du
- Department of Medical Equipment, Wuhan Hospital of Traditional Chinese and Western Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Dabing Huang
- Department of Oncology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Chunguang Miao
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei 230027, China
| | - Bensheng Qiu
- School of Information Science and Technology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Weiping Ding
- Department of Oncology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Chengpan Li
- School of Information Science and Technology, University of Science and Technology of China, Hefei, Anhui 230027, China
- Department of Oncology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| |
Collapse
|
2
|
Du K, Li S, Li C, Li P, Miao C, Luo T, Qiu B, Ding W. Modeling nonalcoholic fatty liver disease on a liver lobule chip with dual blood supply. Acta Biomater 2021; 134:228-239. [PMID: 34265474 DOI: 10.1016/j.actbio.2021.07.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 06/21/2021] [Accepted: 07/06/2021] [Indexed: 12/12/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has emerged as a public health concern. To date, the mechanism of NAFLD progression remains unclear, and pharmacological treatment options are scarce. Traditional animal NAFLD models are limited in helping address these problems due to interspecies differences. Liver chips are promising for modeling NAFLD. However, pre-existing liver chips cannot reproduce complex physicochemical microenvironments of the liver effectively; thus, NAFLD modeling based on these chips is incomplete. Herein, we develop a biomimetic liver lobule chip (LC) and then establish a more accurate on-chip NAFLD model. The self-developed LC achieves dual blood supply through the designed hepatic portal vein and hepatic artery and the microtissue cultured on the LC forms multiple structures similar to in vivo liver. Based on the LC, NAFLD is modeled. Steatosis is successfully induced and more importantly, changing lipid zonation in a liver lobule with the progression of NAFLD is demonstrated for the first time on a microfluidic chip. In addition, the application of the induced NAFLD model has been preliminarily demonstrated in the prevention and reversibility of promising drugs. This study provides a promising platform to understand NAFLD progression and identify drugs for treating NAFLD. STATEMENT OF SIGNIFICANCE: Liver chips are promising for modeling nonalcoholic fatty liver disease. However, on-chip replicating liver physicochemical microenvironments is still a challenge. Herein, we developed a liver lobule chip with dual blood supply, achieving self-organized liver microtissue that is similar to in vivo tissue. Based on the chip, we successfully modeled NAFLD under physiologically differentiated nutrient supplies. For the first time, the changing lipid zonation in a single liver lobule with the early-stage progression of NAFLD was demonstrated on a liver chip. This study provides a promising platform for modeling liver-related diseases.
Collapse
Affiliation(s)
- Kun Du
- Center for Biomedical Engineering, University of Science and Technology of China, Hefei, 230027, China
| | - Shibo Li
- Center for Biomedical Engineering, University of Science and Technology of China, Hefei, 230027, China
| | - Chengpan Li
- Center for Biomedical Engineering, University of Science and Technology of China, Hefei, 230027, China
| | - Ping Li
- Chinese Integrative Medicine Oncology Department, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Chunguang Miao
- Center for Biomedical Engineering, University of Science and Technology of China, Hefei, 230027, China
| | - Tianzhi Luo
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, 230027, China
| | - Bensheng Qiu
- Center for Biomedical Engineering, University of Science and Technology of China, Hefei, 230027, China.
| | - Weiping Ding
- Center for Biomedical Engineering, University of Science and Technology of China, Hefei, 230027, China.
| |
Collapse
|
3
|
Li W, Li P, Li N, Du Y, Lü S, Elad D, Long M. Matrix stiffness and shear stresses modulate hepatocyte functions in a fibrotic liver sinusoidal model. Am J Physiol Gastrointest Liver Physiol 2021; 320:G272-G282. [PMID: 33296275 PMCID: PMC8609567 DOI: 10.1152/ajpgi.00379.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Extracellular matrix (ECM) rigidity has important effects on cell behaviors and increases sharply in liver fibrosis and cirrhosis. Hepatic blood flow is essential in maintaining hepatocytes' (HCs) functions. However, it is still unclear how matrix stiffness and shear stresses orchestrate HC phenotype in concert. A fibrotic three-dimensional (3-D) liver sinusoidal model is constructed using a porous membrane sandwiched between two polydimethylsiloxane (PDMS) layers with respective flow channels. The HCs are cultured in collagen gels of various stiffnesses in the lower channel, whereas the upper channel is pre-seeded with liver sinusoidal endothelial cells (LSECs) and accessible to shear flow. The results reveal that HCs cultured within stiffer matrices exhibit reduced albumin production and cytochrome P450 (CYP450) reductase expression. Low shear stresses enhance synthetic and metabolic functions of HC, whereas high shear stresses lead to the loss of HC phenotype. Furthermore, both mechanical factors regulate HC functions by complementing each other. These observations are likely attributed to mechanically induced mass transport or key signaling molecule of hepatocyte nuclear factor 4α (HNF4α). The present study results provide an insight into understanding the mechanisms of HC dysfunction in liver fibrosis and cirrhosis, especially from the viewpoint of matrix stiffness and blood flow.NEW & NOTEWORTHY A fibrotic three-dimensional (3-D) liver sinusoidal model was constructed to mimic different stages of liver fibrosis in vivo and to explore the cooperative effects of matrix stiffness and shear stresses on hepatocyte (HC) functions. Mechanically induced alterations of mass transport mainly contributed to HC functions via typical mechanosensitive signaling.
Collapse
Affiliation(s)
- Wang Li
- 1Center for Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,2Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,3Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,4School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Peiwen Li
- 1Center for Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,2Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,3Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,4School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Ning Li
- 1Center for Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,2Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,3Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,4School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Yu Du
- 1Center for Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,2Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,3Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,4School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Shouqin Lü
- 1Center for Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,2Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,3Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,4School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - David Elad
- 5Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Mian Long
- 1Center for Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,2Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,3Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,4School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| |
Collapse
|
4
|
Mao Q, Wang L, Liang Y, Dong G, Xia W, Hu J, Xu L, Jiang F. CYP3A5 suppresses metastasis of lung adenocarcinoma through ATOH8/Smad1 axis. Am J Cancer Res 2020; 10:3194-3211. [PMID: 33163265 PMCID: PMC7642649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 08/03/2020] [Indexed: 06/11/2023] Open
Abstract
Cytochrome P450 3A5 (CYP3A5) maintains primary roles in toxic metabolism, catalyzes redox reaction, and contributes to chemotherapeutic resistance. However, the mechanism of CYP3A5 in carcinogenesis remains largely undefined. Here, we investigated a novel role of CYP3A5 inhibiting the metastasis in lung adenocarcinoma (LUAD) via ATOH8/Smad1 axis. We found that CYP3A5 was generally down-regulated in LUAD by RT-PCR, western blot and immunohistochmeistry (IHC) in tissues and cell lines. Low expression of CYP3A5 was significantly associated with poor prognosis of LUAD patients. Functionally, ectopic expression of CYP3A5 could substantially inhibit the migration and invasion in vitro. Consistently, up-regulation of CYP3A5 dramatically suppressed metastatic ability in vivo. Mechanistically, high-throughput phosphorylation chip indicated that CYP3A5 significantly decreased the phosphorylation of Smad1, resulting in suppression of metastasis. Furthermore, bioinformatics analysis and co-immunoprecipitation (Co-IP) experiments uncovered that CYP3A5 interacted with ATOH8, and the interaction, in turn, mediated in-activation in the Smad1 pathway. The combined IHC panel, including CYP3A5 and phosphorylation of Smad1, exhibited a better prognostic value for LUAD patients than any of these components individually. Taken together, CYP3A5 repressed activation of Smad1 to inhibit LUAD metastasis via interacting with ATOH8, indicating a novel potential mechanism of CYP3A5 in LUAD progression.
Collapse
Affiliation(s)
- Qixing Mao
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer HospitalNanjing 210009, P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer HospitalNanjing 210009, P. R. China
| | - Lin Wang
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer HospitalNanjing 210009, P. R. China
- Department of Oncology, Department of Geriatric Lung Cancer Laboratory, The Affiliated Geriatric Hospital of Nanjing Medical UniversityNanjing 210009, P. R. China
| | - Yingkuan Liang
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer HospitalNanjing 210009, P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer HospitalNanjing 210009, P. R. China
| | - Gaochao Dong
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer HospitalNanjing 210009, P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer HospitalNanjing 210009, P. R. China
| | - Wenjie Xia
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer HospitalNanjing 210009, P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer HospitalNanjing 210009, P. R. China
| | - Jianzhong Hu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount SinaiNew York, New York, USA
| | - Lin Xu
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer HospitalNanjing 210009, P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer HospitalNanjing 210009, P. R. China
| | - Feng Jiang
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer HospitalNanjing 210009, P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer HospitalNanjing 210009, P. R. China
| |
Collapse
|
5
|
van der Mark VA, Adam AAA, Chang JC, Oude Elferink RP, Chamuleau RAFM, Hoekstra R. Overexpression of the constitutive androstane receptor and shaken 3D-culturing increase biotransformation and oxidative phosphorylation and sensitivity to mitochondrial amiodarone toxicity of HepaRG cells. Toxicol Appl Pharmacol 2020; 399:115055. [PMID: 32428594 DOI: 10.1016/j.taap.2020.115055] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 05/11/2020] [Accepted: 05/13/2020] [Indexed: 02/06/2023]
Abstract
The liver cell line HepaRG is one of the preferred sources of human hepatocytes for in vitro applications. However, mitochondrial energy metabolism is relatively low, which affects hepatic functionality and sensitivity to hepatotoxins. Culturing in a bioartificial liver (BAL) system with high oxygen, medium perfusion, low substrate stiffness, and 3D conformation increases HepaRG functionality and mitochondrial activity compared to conventional monolayer culturing. In addition, drug metabolism has been improved by overexpression of the constitutive androstane receptor (CAR), a regulator of drug and energy metabolism in the new HepaRG-CAR line. Here, we investigated the effect of BAL culturing on the HepaRG-CAR line by applying a simple and downscaled BAL culture procedure based on shaking 3D cultures, named Bal-in-a-dish (BALIAD). We compared monolayer and BALIAD cultures of HepaRG and HepaRG-CAR cells. CAR overexpression and BALIAD culturing synergistically or additively increased transcript levels of CAR and three of the seven tested CAR target genes in biotransformation. Additionally, Cytochrome P450 3A4 activity was 35-fold increased. The mitochondrial energy metabolism was enhanced; lactate production and glucose consumption switched into lactate elimination and glucose production. BALIAD culturing alone reduced glycogen content and increased oxygen consumption and mitochondrial content. Both CAR overexpression and BALIAD culturing decreased mitochondrial superoxide levels. HepaRG-CAR BALIADs were most sensitive to mitochondrial toxicity induced by the hepatotoxin amiodarone, as indicated by oxygen consumption and mitochondrial superoxide accumulation. These data show that BALIAD culturing of HepaRG-CAR cells induces high mitochondrial energy metabolism and xenobiotic metabolism, increasing its potential for drug toxicity studies.
Collapse
Affiliation(s)
- Vincent A van der Mark
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, AG&M, Meibergdreef 69-71, 1105 BK, Amsterdam, the Netherlands
| | - Aziza A A Adam
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, AG&M, Meibergdreef 69-71, 1105 BK, Amsterdam, the Netherlands.
| | - Jung-Chin Chang
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, AG&M, Meibergdreef 69-71, 1105 BK, Amsterdam, the Netherlands.
| | - Ronald P Oude Elferink
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, AG&M, Meibergdreef 69-71, 1105 BK, Amsterdam, the Netherlands.
| | - Robert A F M Chamuleau
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, AG&M, Meibergdreef 69-71, 1105 BK, Amsterdam, the Netherlands.
| | - Ruurdtje Hoekstra
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, AG&M, Meibergdreef 69-71, 1105 BK, Amsterdam, the Netherlands.
| |
Collapse
|
6
|
Schneeberger K, Sánchez‐Romero N, Ye S, van Steenbeek FG, Oosterhoff LA, Pla Palacin I, Chen C, van Wolferen ME, van Tienderen G, Lieshout R, Colemonts‐Vroninks H, Schene I, Hoekstra R, Verstegen MM, van der Laan LJ, Penning LC, Fuchs SA, Clevers H, De Kock J, Baptista PM, Spee B. Large-Scale Production of LGR5-Positive Bipotential Human Liver Stem Cells. Hepatology 2020; 72:257-270. [PMID: 31715015 PMCID: PMC7496924 DOI: 10.1002/hep.31037] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 11/07/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND AIMS The gap between patients on transplant waiting lists and available donor organs is steadily increasing. Human organoids derived from leucine-rich repeat-containing G protein-coupled receptor 5 (LGR5)-positive adult stem cells represent an exciting new cell source for liver regeneration; however, culturing large numbers of organoids with current protocols is tedious and the level of hepatic differentiation is limited. APPROACH AND RESULTS Here, we established a method for the expansion of large quantities of human liver organoids in spinner flasks. Due to improved oxygenation in the spinner flasks, organoids rapidly proliferated and reached an average 40-fold cell expansion after 2 weeks, compared with 6-fold expansion in static cultures. The organoids repopulated decellularized liver discs and formed liver-like tissue. After differentiation in spinner flasks, mature hepatocyte markers were highly up-regulated compared with static organoid cultures, and cytochrome p450 activity reached levels equivalent to hepatocytes. CONCLUSIONS We established a highly efficient method for culturing large numbers of LGR5-positive stem cells in the form of organoids, which paves the way for the application of organoids for tissue engineering and liver transplantation.
Collapse
Affiliation(s)
- Kerstin Schneeberger
- Department of Clinical Sciences of Companion AnimalsFaculty of Veterinary MedicineUtrecht UniversityUtrechtthe Netherlands
| | | | - Shicheng Ye
- Department of Clinical Sciences of Companion AnimalsFaculty of Veterinary MedicineUtrecht UniversityUtrechtthe Netherlands
| | - Frank G. van Steenbeek
- Department of Clinical Sciences of Companion AnimalsFaculty of Veterinary MedicineUtrecht UniversityUtrechtthe Netherlands
| | - Loes A. Oosterhoff
- Department of Clinical Sciences of Companion AnimalsFaculty of Veterinary MedicineUtrecht UniversityUtrechtthe Netherlands
| | - Iris Pla Palacin
- Instituto de Investigación Sanitaria Aragón (IIS Aragón)ZaragozaSpain
| | - Chen Chen
- Department of Clinical Sciences of Companion AnimalsFaculty of Veterinary MedicineUtrecht UniversityUtrechtthe Netherlands,Hubrecht InstituteRoyal Netherlands Academy of Arts and Sciences and University Medical Center UtrechtUtrechtthe Netherlands
| | - Monique E. van Wolferen
- Department of Clinical Sciences of Companion AnimalsFaculty of Veterinary MedicineUtrecht UniversityUtrechtthe Netherlands
| | - Gilles van Tienderen
- Department of Clinical Sciences of Companion AnimalsFaculty of Veterinary MedicineUtrecht UniversityUtrechtthe Netherlands
| | - Ruby Lieshout
- Department of SurgeryErasmus MC‐University Medical CenterRotterdamthe Netherlands
| | - Haaike Colemonts‐Vroninks
- Department of In Vitro Toxicology and Dermato‐cosmetologyFaculty of Medicine and PharmacyVrije Universiteit BrusselBrusselsBelgium
| | - Imre Schene
- Division of Pediatric GastroenterologyWilhelmina Children's HospitalUniversity Medical Center UtrechtUtrechtthe Netherlands
| | - Ruurdtje Hoekstra
- Tytgat Institute for Liver and Intestinal ResearchGastroenterology and MetabolismAcademic Medical CenterUniversity of AmsterdamAmsterdamthe Netherlands,Surgical LaboratoryDepartment of SurgeryAcademic Medical CenterUniversity of AmsterdamAmsterdamthe Netherlands
| | | | | | - Louis C. Penning
- Department of Clinical Sciences of Companion AnimalsFaculty of Veterinary MedicineUtrecht UniversityUtrechtthe Netherlands
| | - Sabine A. Fuchs
- Division of Pediatric GastroenterologyWilhelmina Children's HospitalUniversity Medical Center UtrechtUtrechtthe Netherlands
| | - Hans Clevers
- Hubrecht InstituteRoyal Netherlands Academy of Arts and Sciences and University Medical Center UtrechtUtrechtthe Netherlands,Cancer Genomics NetherlandsUniversity Medical Center UtrechtUtrechtthe Netherlands,Princess Máxima CenterUtrechtthe Netherlands
| | - Joery De Kock
- Department of In Vitro Toxicology and Dermato‐cosmetologyFaculty of Medicine and PharmacyVrije Universiteit BrusselBrusselsBelgium
| | - Pedro M. Baptista
- Instituto de Investigación Sanitaria Aragón (IIS Aragón)ZaragozaSpain,Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas (CIBERehd)MadridSpain,Fundación ARAIDZaragozaSpain,Instituto de Investigación Sanitaria de la Fundación Jiménez DíazMadridSpain,Department of Biomedical and Aerospace EngineeringUniversidad Carlos III de MadridMadridSpain
| | - Bart Spee
- Department of Clinical Sciences of Companion AnimalsFaculty of Veterinary MedicineUtrecht UniversityUtrechtthe Netherlands
| |
Collapse
|
7
|
Adam AAA, Jongejan A, Moerland PD, van der Mark VA, Oude Elferink RP, Chamuleau RAFM, Hoekstra R. Genome-wide expression profiling reveals increased stability and mitochondrial energy metabolism of the human liver cell line HepaRG-CAR. Cytotechnology 2020; 72:377-395. [PMID: 32130581 PMCID: PMC7225227 DOI: 10.1007/s10616-020-00384-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 02/24/2020] [Indexed: 02/06/2023] Open
Abstract
Human liver cell line HepaRG is a well-known source of human hepatocyte-like cells which, however, displays limited biotransformation and a tendency to transform after 20 passages. The new HepaRG-CAR cell line overexpressing constitutive androstane receptor (CAR, NR1I3), a regulator of detoxification and energy metabolism outperforms the parental HepaRG cell line in various liver functions. To further characterize this cell line and assess its stability we compared HepaRG-CAR with HepaRG cells at different passages for their expression profile, ammonia and lactate metabolism, bile acid and reactive oxygen species (ROS) production. Transcriptomic profiling of HepaRG-CAR vs. HepaRG early-passage revealed downregulation of hypoxia, glycolysis and proliferation and upregulation of oxidative phosphorylation genesets. In addition CAR overexpression downregulated the mTORC1 signaling pathway, which, as mediator of proliferation and metabolic reprogramming, may play an important role in the establishment of the HepaRG-CAR phenotype. The ammonia and lactate metabolism and bile acid production of HepaRG-CAR cells was stable for 10 additional passages compared to HepaRG cells. Interestingly, bile acid production was 4.5-fold higher in HepaRG-CAR vs. HepaRG cells, whereas lactate and ROS production were 2.7- and 2.0-fold lower, respectively. Principal component analysis showed clustering of HepaRG-CAR (early- and late-passage) and HepaRG early-passage and not with HepaRG late-passage indicating that passaging exerted larger effect on the transcriptional profile of HepaRG than HepaRG-CAR cells. In conclusion, overexpression of CAR in HepaRG cells improves their bile acid production, mitochondrial energy metabolism, and stability, with the latter possibly due to reduced ROS production, resulting in an optimized source of human hepatocytes.
Collapse
Affiliation(s)
- Aziza A. A. Adam
- Tytgat Institute for Liver and Intestinal Research, AG&M, Amsterdam UMC, University of Amsterdam, Meibergdreef 69-71, 1105 BK Amsterdam, The Netherlands
| | - Aldo Jongejan
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Perry D. Moerland
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Vincent A. van der Mark
- Tytgat Institute for Liver and Intestinal Research, AG&M, Amsterdam UMC, University of Amsterdam, Meibergdreef 69-71, 1105 BK Amsterdam, The Netherlands
- Surgical Laboratory, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Ronald P. Oude Elferink
- Tytgat Institute for Liver and Intestinal Research, AG&M, Amsterdam UMC, University of Amsterdam, Meibergdreef 69-71, 1105 BK Amsterdam, The Netherlands
| | - Robert A. F. M. Chamuleau
- Tytgat Institute for Liver and Intestinal Research, AG&M, Amsterdam UMC, University of Amsterdam, Meibergdreef 69-71, 1105 BK Amsterdam, The Netherlands
| | - Ruurdtje Hoekstra
- Tytgat Institute for Liver and Intestinal Research, AG&M, Amsterdam UMC, University of Amsterdam, Meibergdreef 69-71, 1105 BK Amsterdam, The Netherlands
- Surgical Laboratory, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
8
|
Pasqua M, Pereira U, Messina A, de Lartigue C, Vigneron P, Dubart-Kupperschmitt A, Legallais C. HepaRG Self-Assembled Spheroids in Alginate Beads Meet the Clinical Needs for Bioartificial Liver. Tissue Eng Part A 2020; 26:613-622. [PMID: 31914890 DOI: 10.1089/ten.tea.2019.0262] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
In liver tissue engineering, cell culture in spheroids is now well recognized to promote the maintenance of hepatic functions. However, the process leading to spheroids formation is time consuming, costly, and not easy to scale-up for further use in human bioartificial liver (BAL) applications. In this study, we encapsulated HepaRG cells (precursors of hepatocyte-like cells) in 1.5% alginate beads without preforming spheroids. Starting from a given hepatic biomass, we analyzed cell differentiation and metabolic performance for further use in a fluidized-bed BAL. We observed that cells self-rearranged as aggregates within the beads and adequately differentiated over time, in the absence of any differentiating factors classically used. On day 14 postencapsulation, cells displayed a wide range of hepatic features necessary for the treatment of a patient in acute liver failure. These activities include albumin synthesis, ammonia and lactate detoxification, and the efficacy of the enzymes involved in the xenobiotic metabolism (such as CYP1A1/2). Impact statement It has been recognized that culturing cells in spheroids (SPHs) is advantageous as they better reproduce the three-dimensional physiological microenvironment. This approach can be exploited in bioartificial liver applications, where obtaining a functional hepatic biomass is the major challenge. Our study describes an original method for culturing hepatic cells in alginate beads that makes possible the autonomous formation of SPHs after 3 days of culture. In turn, the cells differentiate adequately and display a wide range of hepatic features. They are also capable of treating a pathological plasma model. Finally, this setup can easily be scaled-up to treat acute liver failure.
Collapse
Affiliation(s)
- Mattia Pasqua
- UMR CNRS 7338 Biomechanics & Bioengineering, Université de Technologie de Compiègne, Alliance Sorbonne Université, Compiègne, France
| | - Ulysse Pereira
- UMR CNRS 7338 Biomechanics & Bioengineering, Université de Technologie de Compiègne, Alliance Sorbonne Université, Compiègne, France
| | - Antonietta Messina
- DHU Hépatinov, Villejuif, France.,UMR_S1193 Inserm/Paris-Saclay University, Villejuif, France
| | - Claire de Lartigue
- UMR CNRS 7338 Biomechanics & Bioengineering, Université de Technologie de Compiègne, Alliance Sorbonne Université, Compiègne, France
| | - Pascale Vigneron
- UMR CNRS 7338 Biomechanics & Bioengineering, Université de Technologie de Compiègne, Alliance Sorbonne Université, Compiègne, France
| | | | - Cecile Legallais
- UMR CNRS 7338 Biomechanics & Bioengineering, Université de Technologie de Compiègne, Alliance Sorbonne Université, Compiègne, France.,DHU Hépatinov, Villejuif, France
| |
Collapse
|
9
|
Lynch MA. Can the emerging field of immunometabolism provide insights into neuroinflammation? Prog Neurobiol 2019; 184:101719. [PMID: 31704314 DOI: 10.1016/j.pneurobio.2019.101719] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 10/18/2019] [Accepted: 10/30/2019] [Indexed: 12/29/2022]
Abstract
In the past few years it has become increasingly clear that an understanding of the interaction between metabolism and immune function can provide an insight into cellular responses to challenges. Significant progress has been made in terms of how macrophages are metabolically re-programmed in response to inflammatory stimuli but, to date, little emphasis has been placed on evaluating equivalent changes in microglia. The need to make progress is driven by the fact that, while microglial activation and the cell's ability to adopt an inflammatory phenotype is necessary to fulfil the neuroprotective function of the cell, persistent activation of microglia and the associated neuroinflammation is at the heart of several neurodegenerative diseases. Understanding the metabolic changes that accompany microglial responses may broaden our perspective on how dysfunction might arise and be tempered. This review will evaluate the current literature that addresses the interplay between inflammation and metabolic reprogramming in microglia, reflecting on the parallels that exist with macrophages. It will consider the changes that take place with age including those that have been reported in neurons and astrocytes with the development of non-invasive imaging techniques, and reflect on the literature that is currently available relating to metabolic reprogramming of microglia with age and in neurodegeneration. Finally it will consider the possibility that manipulating microglial metabolism may provide a valuable approach to modulating neuroinflammation.
Collapse
Affiliation(s)
- Marina A Lynch
- Trinity College Institute of Neuroscience, Trinity College, Dublin 2, Ireland.
| |
Collapse
|
10
|
Chamuleau RAFM, Hoekstra R. End-stage liver failure: filling the treatment gap at the intensive care unit. J Artif Organs 2019; 23:113-123. [PMID: 31535298 PMCID: PMC7228976 DOI: 10.1007/s10047-019-01133-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 09/09/2019] [Indexed: 12/14/2022]
Abstract
End-stage liver failure is a condition of collapsing liver function with mortality rates up to 80. Liver transplantation is the only lifesaving therapy. There is an unmet need for therapy to extend the waiting time for liver transplantation or regeneration of the native liver. Here we review the state-of-the-art of non-cell based and cell-based artificial liver support systems, cell transplantation and plasma exchange, with the first therapy relying on detoxification, while the others aim to correct also other failing liver functions and/or modulate the immune response. Meta-analyses on the effect of non-cell based systems show contradictory outcomes for different types of albumin purification devices. For bioartificial livers proof of concept has been shown in animals with liver failure. However, large clinical trials with two different systems did not show a survival benefit. Two clinical trials with plasma exchange and one with transplantation of mesenchymal stem cells showed positive outcomes on survival. Detoxification therapies lack adequacy for most patients. Correction of additional liver functions, and also modulation of the immune system hold promise for future therapy of liver failure.
Collapse
Affiliation(s)
- Robert A F M Chamuleau
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, AG&M, Academic Medical Center, Meibergdreef 69-71, S1-176, 1105 BK, Amsterdam, The Netherlands.
| | - Ruurdtje Hoekstra
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, AG&M, Academic Medical Center, Meibergdreef 69-71, S1-176, 1105 BK, Amsterdam, The Netherlands
| |
Collapse
|
11
|
Adam AAA, van der Mark VA, Ruiter JPN, Wanders RJA, Oude Elferink RPJ, Chamuleau RAFM, Hoekstra R. Overexpression of carbamoyl-phosphate synthase 1 significantly improves ureagenesis of human liver HepaRG cells only when cultured under shaking conditions. Mitochondrion 2019; 47:298-308. [PMID: 30802674 DOI: 10.1016/j.mito.2019.02.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 01/17/2019] [Accepted: 02/21/2019] [Indexed: 12/12/2022]
Abstract
Hyperammonemia is an important contributing factor to hepatic encephalopathy in end-stage liver failure patients. Therefore reducing hyperammonemia is a requisite of bioartificial liver support (BAL). Ammonia elimination by human liver HepaRG cells occurs predominantly through reversible fixation into amino acids, whereas the irreversible conversion into urea is limited. Compared to human liver, the expression and activity of the three urea cycle (UC) enzymes carbamoyl-phosphate synthase1 (CPS1), ornithine transcarbamoylase (OTC) and arginase1, are low. To improve HepaRG cells as BAL biocomponent, its rate limiting factor of the UC was determined under two culture conditions: static and dynamic medium flow (DMF) achieved by shaking. HepaRG cells increasingly converted escalating arginine doses into urea, indicating that arginase activity is not limiting ureagenesis. Neither was OTC activity, as a stable HepaRG line overexpressing OTC exhibited a 90- and 15.7-fold upregulation of OTC transcript and activity levels, without improvement in ureagenesis. However, a stable HepaRG line overexpressing CPS1 showed increased mitochondrial stress and reduced hepatic differentiation without promotion of the CPS1 transcript level or ureagenesis under static-culturing conditions, yet, it exhibited a 4.3-fold increased ureagenesis under DMF. This was associated with increased CPS1 transcript and activity levels amounting to >2-fold, increased mitochondrial abundance and hepatic differentiation. Unexpectedly, the transcript levels of several other UC genes increased up to 6.8-fold. We conclude that ureagenesis can be improved in HepaRG cells by CPS1 overexpression, however, only in combination with DMF-culturing, suggesting that both the low CPS1 level and static-culturing, possibly due to insufficient mitochondria, are limiting UC.
Collapse
Affiliation(s)
- Aziza A A Adam
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, AG&M, Meibergdreef 69-71, 1105 BK Amsterdam, The Netherlands
| | - Vincent A van der Mark
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, AG&M, Meibergdreef 69-71, 1105 BK Amsterdam, The Netherlands; Amsterdam UMC, University of Amsterdam, Surgical Laboratory, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Jos P N Ruiter
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands
| | - Ronald J A Wanders
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands
| | - Ronald P J Oude Elferink
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, AG&M, Meibergdreef 69-71, 1105 BK Amsterdam, The Netherlands
| | - Robert A F M Chamuleau
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, AG&M, Meibergdreef 69-71, 1105 BK Amsterdam, The Netherlands
| | - Ruurdtje Hoekstra
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, AG&M, Meibergdreef 69-71, 1105 BK Amsterdam, The Netherlands; Amsterdam UMC, University of Amsterdam, Surgical Laboratory, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands.
| |
Collapse
|