1
|
Miura S, Horisawa K, Iwamori T, Tsujino S, Inoue K, Karasawa S, Yamamoto J, Ohkawa Y, Sekiya S, Suzuki A. Hepatocytes differentiate into intestinal epithelial cells through a hybrid epithelial/mesenchymal cell state in culture. Nat Commun 2024; 15:3940. [PMID: 38750036 PMCID: PMC11096382 DOI: 10.1038/s41467-024-47869-2] [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: 01/15/2023] [Accepted: 04/14/2024] [Indexed: 05/18/2024] Open
Abstract
Hepatocytes play important roles in the liver, but in culture, they immediately lose function and dedifferentiate into progenitor-like cells. Although this unique feature is well-known, the dynamics and mechanisms of hepatocyte dedifferentiation and the differentiation potential of dedifferentiated hepatocytes (dediHeps) require further investigation. Here, we employ a culture system specifically established for hepatic progenitor cells to study hepatocyte dedifferentiation. We found that hepatocytes dedifferentiate with a hybrid epithelial/mesenchymal phenotype, which is required for the induction and maintenance of dediHeps, and exhibit Vimentin-dependent propagation, upon inhibition of the Hippo signaling pathway. The dediHeps re-differentiate into mature hepatocytes by forming aggregates, enabling reconstitution of hepatic tissues in vivo. Moreover, dediHeps have an unexpected differentiation potential into intestinal epithelial cells that can form organoids in three-dimensional culture and reconstitute colonic epithelia after transplantation. This remarkable plasticity will be useful in the study and treatment of intestinal metaplasia and related diseases in the liver.
Collapse
Affiliation(s)
- Shizuka Miura
- Division of Organogenesis and Regeneration, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Kenichi Horisawa
- Division of Organogenesis and Regeneration, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Tokuko Iwamori
- Division of Organogenesis and Regeneration, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Satoshi Tsujino
- Division of Organogenesis and Regeneration, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Kazuya Inoue
- Division of Organogenesis and Regeneration, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Satsuki Karasawa
- Division of Organogenesis and Regeneration, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Junpei Yamamoto
- Division of Organogenesis and Regeneration, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Sayaka Sekiya
- Division of Organogenesis and Regeneration, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Atsushi Suzuki
- Division of Organogenesis and Regeneration, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan.
| |
Collapse
|
2
|
Andres S, Bartling B, Stiensmeier V, Starke A, Schmicke M. Comparative cryopreservation of bovine and porcine primary hepatocytes. Front Vet Sci 2023; 10:1211135. [PMID: 37614462 PMCID: PMC10442649 DOI: 10.3389/fvets.2023.1211135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 06/09/2023] [Indexed: 08/25/2023] Open
Abstract
The isolation of primary hepatocytes from liver tissue of farm animals yields a very high number of cells, and a part of them can be stored by cryopreservation for future experiments. As no experience exists with the cryopreservation of hepatocytes from cattle, our study aimed at the cryopreservation of bovine hepatocytes by use of different protocols compared with the cryopreservation of hepatocytes from pig. We tested different freezing media (William's Medium E vs. University of Wisconsin solution), cryoprotectants (dimethyl sulfoxide with vs. without trehalose as additional additive), freezing systems (standard freezing container vs. controlled-rate freezer) and freezing times (4 vs. 28 d). These tests identified a general influence of species and freezing systems, whereas the influence of freezing media, trehalose additive and freezing time was less or not obvious. In this regard, we determined a mean recovery of 30% of bovine hepatocytes and 55% of porcine hepatocytes cryopreserved in a controlled-rate freezer, whereas the rates were about 10% less when hepatocytes were frozen in a standard freezing container. In accordance with this observation, the cultivation of cryopreserved hepatocytes from cattle was less effective than that of porcine hepatocytes. Hepatocytes from cattle can be successfully cryopreserved and partially cultured after cryopreservation but with lower percentage than porcine hepatocytes.
Collapse
Affiliation(s)
- Sandra Andres
- Institute of Agricultural and Nutritional Sciences, Animal Health Management, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Babett Bartling
- Institute of Agricultural and Nutritional Sciences, Animal Health Management, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Vera Stiensmeier
- Institute of Agricultural and Nutritional Sciences, Animal Health Management, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Alexander Starke
- Department for Ruminants and Swine, Faculty of Veterinary Medicine, Leipzig University, Leipzig, Germany
| | - Marion Schmicke
- Institute of Agricultural and Nutritional Sciences, Animal Health Management, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
- Clinic for Cattle, Endocrinology, University of Veterinary Medicine Hannover, Hanover, Germany
| |
Collapse
|
3
|
Singh V, Lall N, Wadhwani A, Dhanabal SP. GC-MS analysis of curculigo orchiodes and medicinal herbs with cytotoxic, hepatoprotective attributes of ethanolic extract from Indian origin. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2022; 19:719-727. [PMID: 35320638 DOI: 10.1515/jcim-2020-0545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 02/27/2022] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Liver illnesses are a major public health issue all over the world. Medicinal plants constituents a viable alternative for the development of phytopharmaceuticals with hepatoprotective activity in order to solve some of these health-related problems. The present study is focused on the phytochemical and biological investigation on Indian traditional medicinal plant extracts, for their cytotoxic and hepatoprotective activity. The isolated compounds showed the presence of phenolic constituents which lead to cytotoxicity and hepatoprotective activity of medicinal plant. Cancer causes about 13% of all human deaths in 2007 (7.6 million) (American Cancer Society and WHO December 2006-07). The American Cancer Society estimates that 12,990 new cases of cervical cancer will be diagnosed in the United States year 2016. Cancer-related deaths are expected to increase, with an estimated 11.4 million deaths in 2030. METHODS The ethanolic extracts of Centella asiatica, Myristica fragrans, Trichosanthes palmata, Woodfordia fruticosa, Curculigo orchioides were evaluated against HEP-G2 cell lines for hepatoprotective activity and Curculigo orchioides was further promoted for the isolation of secondary metabolites based on inhibitory concentration. RESULTS The ethanolic extracts of C. asiatica, M. fragrans, T. palmata, W. fruticosa, Curculigo orchioides shown significant cytotoxic activity (IC50≤100 μg/mL). The plant extracts also shown significant hepatoprotective activity in a dose dependent manner when tested against HEP-G2 cell lines and cytotoxicity studies against HeLa and HEP-G2 cells. CONCLUSIONS The extract of Curculigo orchiodes rhizome showed significant cytotoxicity results. Hence the Curculigo orchiodes rhizome was selected for further phytochemical studies to isolate active compounds and their Characterization by GCMS.
Collapse
Affiliation(s)
- Vedpal Singh
- Department of Pharmacognosy, College of Pharmacy, JSS Academy of Technical Education, Noida, UP, India
| | - Namrita Lall
- Department of Plant and Soil Sciences, University of Pretoria, Pretoria, South Africa
| | - Ashish Wadhwani
- Department of Pharmaceutical Biotechnology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, Tamilnadu, India.,School of Pharmacy, Faculty of Health Science, JSS Academy of Higher Education and Research, Mauritius campus. Droopnath Ramphul St, Vacoas-Phoenix, Mauritius
| | - Sangai P Dhanabal
- Department of Pharmacognosy, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Tamilnadu, India
| |
Collapse
|
4
|
Lee C, Kim J, Han J, Oh D, Kim M, Jeong H, Kim TJ, Kim SW, Kim JN, Seo YS, Suzuki A, Kim JH, Jung Y. Formyl peptide receptor 2 determines sex-specific differences in the progression of nonalcoholic fatty liver disease and steatohepatitis. Nat Commun 2022; 13:578. [PMID: 35102146 PMCID: PMC8803937 DOI: 10.1038/s41467-022-28138-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/12/2022] [Indexed: 12/21/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is an important health concern worldwide and progresses into nonalcoholic steatohepatitis (NASH). Although prevalence and severity of NAFLD/NASH are higher in men than premenopausal women, it remains unclear how sex affects NAFLD/NASH pathophysiology. Formyl peptide receptor 2 (FPR2) modulates inflammatory responses in several organs; however, its role in the liver is unknown. Here we show that FPR2 mediates sex-specific responses to diet-induced NAFLD/NASH. NASH-like liver injury was induced in both sexes during choline-deficient, L-amino acid-defined, high-fat diet (CDAHFD) feeding, but compared with females, male mice had more severe hepatic damage. Fpr2 was more highly expressed in hepatocytes and healthy livers from females than males, and FPR2 deletion exacerbated liver damage in CDAHFD-fed female mice. Estradiol induced Fpr2 expression, which protected hepatocytes and the liver from damage. In conclusion, our results demonstrate that FPR2 mediates sex-specific responses to diet-induced NAFLD/NASH, suggesting a novel therapeutic target for NAFLD/NASH.
Collapse
Affiliation(s)
- Chanbin Lee
- Department of Integrated Biological Science, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea
| | - Jieun Kim
- Department of Integrated Biological Science, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea
| | - Jinsol Han
- Department of Integrated Biological Science, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea
| | - Dayoung Oh
- Department of Integrated Biological Science, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea
| | - Minju Kim
- Department of Integrated Biological Science, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea
| | - Hayeong Jeong
- Department of Integrated Biological Science, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea
| | - Tae-Jin Kim
- Department of Integrated Biological Science, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea
- Department of Biological Sciences, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea
| | - Sang-Woo Kim
- Department of Integrated Biological Science, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea
- Department of Biological Sciences, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea
| | - Jeong Nam Kim
- Department of Integrated Biological Science, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea
- Department of Microbiology, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea
| | - Young-Su Seo
- Department of Integrated Biological Science, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea
- Department of Microbiology, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea
| | - Ayako Suzuki
- Division of Gastroenterology and Hepatology, Duke University, Durham, NC, USA
| | - Jae Ho Kim
- Department of Physiology, Pusan National University School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Youngmi Jung
- Department of Integrated Biological Science, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea.
- Department of Biological Sciences, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea.
| |
Collapse
|
5
|
Horisawa K, Udono M, Ueno K, Ohkawa Y, Nagasaki M, Sekiya S, Suzuki A. The Dynamics of Transcriptional Activation by Hepatic Reprogramming Factors. Mol Cell 2020; 79:660-676.e8. [PMID: 32755593 DOI: 10.1016/j.molcel.2020.07.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/18/2020] [Accepted: 07/09/2020] [Indexed: 02/07/2023]
Abstract
Specific combinations of two transcription factors (Hnf4α plus Foxa1, Foxa2, or Foxa3) can induce direct conversion of mouse fibroblasts into hepatocyte-like cells. However, the molecular mechanisms underlying hepatic reprogramming are largely unknown. Here, we show that the Foxa protein family members and Hnf4α sequentially and cooperatively bind to chromatin to activate liver-specific gene expression. Although all Foxa proteins bind to and open regions of closed chromatin as pioneer factors, Foxa3 has the unique potential of transferring from the distal to proximal regions of the transcription start site of target genes, binding RNA polymerase II, and co-traversing target genes. These distinctive characteristics of Foxa3 are essential for inducing the hepatic fate in fibroblasts. Similar functional coupling of transcription factors to RNA polymerase II may occur in other contexts whereby transcriptional activation can induce cell differentiation.
Collapse
Affiliation(s)
- Kenichi Horisawa
- Division of Organogenesis and Regeneration, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Miyako Udono
- Division of Organogenesis and Regeneration, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Kazuko Ueno
- Genome Medical Science Project, National Center for Global Health and Medicine, Tokyo 162-8655, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Masao Nagasaki
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan; Human Biosciences Unit for the Top Global Course, Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto 606-8507, Japan
| | - Sayaka Sekiya
- Division of Organogenesis and Regeneration, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Atsushi Suzuki
- Division of Organogenesis and Regeneration, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan.
| |
Collapse
|
6
|
Hong T, Ge Z, Zhang B, Meng R, Zhu D, Bi Y. Erythropoietin suppresses hepatic steatosis and obesity by inhibiting endoplasmic reticulum stress and upregulating fibroblast growth factor 21. Int J Mol Med 2019; 44:469-478. [PMID: 31173165 PMCID: PMC6605699 DOI: 10.3892/ijmm.2019.4210] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 05/23/2019] [Indexed: 12/13/2022] Open
Abstract
Erythropoietin (EPO), known primarily for its role in erythropoiesis, was recently reported to play a beneficial role in regulating lipid metabolism; however, the underlying mechanism through which EPO decreases hepatic lipid accumulation requires further investigation. Endoplasmic reticulum (ER) stress may contribute to the progression of hepatic steatosis. The present study investigated the effects of EPO on regulating ER stress in fatty liver. It was demonstrated that EPO inhibited hepatic ER stress and steatosis in vivo and in vitro. Interestingly, these beneficial effects were abrogated in liver-specific sirtuin 1 (SIRT1)-knockout mice compared with wild-type littermates. In addition, in palmitate-treated hepatocytes, small interfering RNA-mediated SIRT1 silencing suppressed the effects of EPO on lipid-induced ER stress. Additionally, EPO stimulated hepatic fibroblast growth factor 21 (FGF21) expression and secretion in a SIRT1-dependent manner in mice. Furthermore, the sensitivity of hepatocytes from obese mice to FGF21 was restored following treatment with EPO. Collectively, the results of the present study revealed a new mechanism underlying the regulation of hepatic ER stress and FGF21 expression induced by EPO; thus, EPO may be considered as a potential therapeutic agent for the treatment of fatty liver and obesity.
Collapse
Affiliation(s)
- Ting Hong
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Zhijuan Ge
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Bingjie Zhang
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Ran Meng
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Dalong Zhu
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Yan Bi
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| |
Collapse
|
7
|
Hong T, Ge Z, Meng R, Wang H, Zhang P, Tang S, Lu J, Gu T, Zhu D, Bi Y. Erythropoietin alleviates hepatic steatosis by activating SIRT1-mediated autophagy. Biochim Biophys Acta Mol Cell Biol Lipids 2018. [PMID: 29522896 DOI: 10.1016/j.bbalip.2018.03.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Erythropoietin (EPO), besides its stimulatory effect on erythropoiesis, is beneficial to insulin resistance and obesity. However, its role in hepatic steatosis remains unexplored. Activating autophagy seems a promising mechanism for improving fatty liver disease. The present study investigated the role of EPO in alleviating hepatic steatosis and sought to determine whether its function is mediated by the activation of autophagy. Here, we show that EPO decreased hepatic lipid content significantly in vivo and in vitro. Furthermore, EPO/EPO receptor (EPOR) signalling induced autophagy activation in hepatocytes as indicated by western blot assay, transmission electron microscopy, and confocal microscopy. In addition, EPO increased the co-localization of autophagosomes and cellular lipids as shown by double labelling of the autophagy marker light chain microtubule-associated protein 3 (LC3) and lipids. Importantly, suppression of autophagy by an inhibitor or small interfering RNA (siRNA) abolished the EPO-mediated alleviation hepatic steatosis in vitro. Furthermore, EPO up-regulated sirtuin 1 (SIRT1) expression, and siRNA-mediated SIRT1 silencing abrogated the EPO-induced increases in LC3 protein and deacetylation levels, thereby preventing the alleviation of hepatic steatosis. Taken together, this study revealed a new mechanism wherein EPO alleviates hepatic steatosis by activating autophagy via SIRT1-dependent deacetylation of LC3. This finding might have therapeutic value in the treatment of hepatic steatosis.
Collapse
Affiliation(s)
- Ting Hong
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, No 321 Zhongshan Road, Nanjing, China
| | - Zhijuan Ge
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, No 321 Zhongshan Road, Nanjing, China
| | - Ran Meng
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, No 321 Zhongshan Road, Nanjing, China
| | - Hongdong Wang
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, No 321 Zhongshan Road, Nanjing, China
| | - Pengzi Zhang
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, No 321 Zhongshan Road, Nanjing, China
| | - Sunyinyan Tang
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, No 321 Zhongshan Road, Nanjing, China
| | - Jing Lu
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, No 321 Zhongshan Road, Nanjing, China
| | - Tianwei Gu
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, No 321 Zhongshan Road, Nanjing, China
| | - Dalong Zhu
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, No 321 Zhongshan Road, Nanjing, China.
| | - Yan Bi
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, No 321 Zhongshan Road, Nanjing, China.
| |
Collapse
|
8
|
Shukla I, Azmi L, Gupta SS, Upreti DK, Rao CV. Amelioration of anti-hepatotoxic effect by Lichen rangiferinus against alcohol induced liver damage in rats. J Ayurveda Integr Med 2018; 10:171-177. [PMID: 29395895 PMCID: PMC6822147 DOI: 10.1016/j.jaim.2017.08.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 07/21/2017] [Accepted: 08/22/2017] [Indexed: 12/20/2022] Open
Abstract
Background Reindeer lichen, Lichen rangiferinus syn. or Cladonia rangiferina (L.) F. H. Wigg. (Cladoniaceae) has been traditionally reported as a remedy to treat fever, colds, arthritis as well as convulsions, liver infections, coughs, constipation, and tuberculosis. The current study is aimed at rectification of alcohol induced liver damage by the use of L. rangiferinus extract. Objectives The aim of the study was to compare some biochemical markers for liver injury and hematological indices in normal untreated rats and treated rats. Material and Methods The study was performed using male Wistar rats. Animals were categorized into five groups, negative control group (normal diet only), treated groups (2 groups were lichen treated along with 10% ethanol & 1 group was only ethanol treated) and positive control group (Silymarin + 10% ethanol) of six animals in each group. Biochemical markers for liver injury and hematological indices of all animals were measured using standard diagnostic tools. The animals were then sacrificed and livers were sent to the pathology lab for histopathological analysis. Results Lichen extract showed a significant restoration of altered biochemical parameters towards normal in both in vitro and in vivo conditions. The total phenolic and flavonoid content of the LRE was found to be 21.78 μg PE/mg of extract and 5.13 μg RE/mg of extract respectively. The IC50 values for atranorin and fumarprotocetraric acid were found to be 128.48 and 218.46 mg/mL respectively. Reducing power of the extract was found to be quite significant. After administration of lichen extract, endothelial cells were less injured around central vein and number of fat vacuoles was also lesser in hepatocytes. Conclusion Conclusively, treatment with lichen extract assuages alcohol-related damage and guards hepatic tissue from alcohol-induced toxicity. Alcohol consumption causes a number of liver ailments which when not corrected leads to alcohol liver disease. The current study is aimed at rectification of alcohol induced liver damage by the use of Lichen rangiferinus in rats. Tested biochemical parameters showed reduction, following administration of lichen extract. Conclusively, lichen extract assuages alcohol-related damage and guards liver from alcohol-induced toxicity.
Collapse
Affiliation(s)
- Ila Shukla
- CSIR - National Botanical Research Institute, Lucknow 226001, Uttar Pradesh, India
| | - Lubna Azmi
- CSIR - National Botanical Research Institute, Lucknow 226001, Uttar Pradesh, India
| | - Shyam Sundar Gupta
- CSIR - National Botanical Research Institute, Lucknow 226001, Uttar Pradesh, India
| | - Dalip Kumar Upreti
- CSIR - National Botanical Research Institute, Lucknow 226001, Uttar Pradesh, India
| | | |
Collapse
|
9
|
Ogoke O, Oluwole J, Parashurama N. Bioengineering considerations in liver regenerative medicine. J Biol Eng 2017; 11:46. [PMID: 29204185 PMCID: PMC5702480 DOI: 10.1186/s13036-017-0081-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 09/25/2017] [Indexed: 12/19/2022] Open
Abstract
Background Liver disease contributes significantly to global disease burden and is associated with rising incidence and escalating costs. It is likely that innovative approaches, arising from the emerging field of liver regenerative medicine, will counter these trends. Main body Liver regenerative medicine is a rapidly expanding field based on a rich history of basic investigations into the nature of liver structure, physiology, development, regeneration, and function. With a bioengineering perspective, we discuss all major subfields within liver regenerative medicine, focusing on the history, seminal publications, recent progress within these fields, and commercialization efforts. The areas reviewed include fundamental aspects of liver transplantation, liver regeneration, primary hepatocyte cell culture, bioartificial liver, hepatocyte transplantation and liver cell therapies, mouse liver repopulation, adult liver stem cell/progenitor cells, pluripotent stem cells, hepatic microdevices, and decellularized liver grafts. Conclusion These studies highlight the creative directions of liver regenerative medicine, the collective efforts of scientists, engineers, and doctors, and the bright outlook for a wide range of approaches and applications which will impact patients with liver disease.
Collapse
Affiliation(s)
- Ogechi Ogoke
- Department of Chemical and Biological Engineering, University at Buffalo (State University of New York), Furnas Hall, Buffalo, NY 14260 USA.,Clinical and Translation Research Center (CTRC), University at Buffalo (State University of New York), 875 Ellicott St., Buffalo, NY 14203 USA
| | - Janet Oluwole
- Clinical and Translation Research Center (CTRC), University at Buffalo (State University of New York), 875 Ellicott St., Buffalo, NY 14203 USA.,Department of Biomedical Engineering, University at Buffalo (State University of New York), Furnas Hall, 907 Furnas Hall, Buffalo, NY 14260 USA
| | - Natesh Parashurama
- Department of Chemical and Biological Engineering, University at Buffalo (State University of New York), Furnas Hall, Buffalo, NY 14260 USA.,Clinical and Translation Research Center (CTRC), University at Buffalo (State University of New York), 875 Ellicott St., Buffalo, NY 14203 USA.,Department of Biomedical Engineering, University at Buffalo (State University of New York), Furnas Hall, 907 Furnas Hall, Buffalo, NY 14260 USA
| |
Collapse
|
10
|
Kim SH, Kim G, Han DH, Lee M, Kim I, Kim B, Kim KH, Song YM, Yoo JE, Wang HJ, Bae SH, Lee YH, Lee BW, Kang ES, Cha BS, Lee MS. Ezetimibe ameliorates steatohepatitis via AMP activated protein kinase-TFEB-mediated activation of autophagy and NLRP3 inflammasome inhibition. Autophagy 2017; 13:1767-1781. [PMID: 28933629 DOI: 10.1080/15548627.2017.1356977] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Impairment in macroautophagy/autophagy flux and inflammasome activation are common characteristics of nonalcoholic steatohepatitis (NASH). Considering the lack of approved agents for treating NASH, drugs that can enhance autophagy and modulate inflammasome pathways may be beneficial. Here, we investigated the novel mechanism of ezetimibe, a widely prescribed drug for hypercholesterolemia, as a therapeutic option for ameliorating NASH. Human liver samples with steatosis and NASH were analyzed. For in vitro studies of autophagy and inflammasomes, primary mouse hepatocytes, human hepatoma cells, mouse embryonic fibroblasts with Ampk or Tsc2 knockout, and human or primary mouse macrophages were treated with ezetimibe and palmitate. Steatohepatitis and fibrosis were induced by feeding Atg7 wild-type, haploinsufficient, and knockout mice a methionine- and choline-deficient diet with ezetimibe (10 mg/kg) for 4 wk. Human livers with steatosis or NASH presented impaired autophagy with decreased nuclear TFEB and increased SQSTM1, MAP1LC3-II, and NLRP3 expression. Ezetimibe increased autophagy flux and concomitantly ameliorated lipid accumulation and apoptosis in palmitate-exposed hepatocytes. Ezetimibe induced AMPK phosphorylation and subsequent TFEB nuclear translocation, related to MAPK/ERK. In macrophages, ezetimibe blocked the NLRP3 inflammasome-IL1B pathway in an autophagy-dependent manner and modulated hepatocyte-macrophage interaction via extracellular vesicles. Ezetimibe attenuated lipid accumulation, inflammation, and fibrosis in liver-specific Atg7 wild-type and haploinsufficient mice, but not in knockout mice. Ezetimibe ameliorates steatohepatitis by autophagy induction through AMPK activation and TFEB nuclear translocation, related to an independent MTOR ameliorative effect and the MAPK/ERK pathway. Ezetimibe dampens NLRP3 inflammasome activation in macrophages by modulating autophagy and a hepatocyte-driven exosome pathway.
Collapse
Affiliation(s)
- Soo Hyun Kim
- a Department of Internal Medicine , Yonsei University College of Medicine , Seoul , Korea
| | - Gyuri Kim
- a Department of Internal Medicine , Yonsei University College of Medicine , Seoul , Korea.,b Department of Medicine, Samsung Medical Center , Sungkyunkwan University School of Medicine , Seoul , Korea.,c Graduate School , Yonsei University College of Medicine , Seoul , Korea
| | - Dai Hoon Han
- d Department of Surgery , Yonsei University College of Medicine , Seoul , Korea
| | - Milim Lee
- a Department of Internal Medicine , Yonsei University College of Medicine , Seoul , Korea
| | - Irene Kim
- a Department of Internal Medicine , Yonsei University College of Medicine , Seoul , Korea
| | - Bohkyung Kim
- a Department of Internal Medicine , Yonsei University College of Medicine , Seoul , Korea
| | - Kook Hwan Kim
- e Severance Biomedical Science Institute, Yonsei Biomedical Research Institute , Yonsei University College of Medicine , Seoul , Korea
| | - Young-Mi Song
- f Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital , University of Toronto , Toronto , Canada
| | - Jeong Eun Yoo
- g Department of Pathology , Yonsei University College of Medicine , Seoul , Korea
| | - Hye Jin Wang
- h Department of Pharmacology , Yonsei University College of Medicine , Seoul , Korea
| | - Soo Han Bae
- e Severance Biomedical Science Institute, Yonsei Biomedical Research Institute , Yonsei University College of Medicine , Seoul , Korea
| | - Yong-Ho Lee
- a Department of Internal Medicine , Yonsei University College of Medicine , Seoul , Korea.,c Graduate School , Yonsei University College of Medicine , Seoul , Korea.,i Institute of Endocrine Research , Yonsei University College of Medicine , Seoul , Korea
| | - Byung-Wan Lee
- a Department of Internal Medicine , Yonsei University College of Medicine , Seoul , Korea.,c Graduate School , Yonsei University College of Medicine , Seoul , Korea.,i Institute of Endocrine Research , Yonsei University College of Medicine , Seoul , Korea
| | - Eun Seok Kang
- a Department of Internal Medicine , Yonsei University College of Medicine , Seoul , Korea.,c Graduate School , Yonsei University College of Medicine , Seoul , Korea.,i Institute of Endocrine Research , Yonsei University College of Medicine , Seoul , Korea
| | - Bong-Soo Cha
- a Department of Internal Medicine , Yonsei University College of Medicine , Seoul , Korea.,c Graduate School , Yonsei University College of Medicine , Seoul , Korea.,i Institute of Endocrine Research , Yonsei University College of Medicine , Seoul , Korea
| | - Myung-Shik Lee
- a Department of Internal Medicine , Yonsei University College of Medicine , Seoul , Korea.,e Severance Biomedical Science Institute, Yonsei Biomedical Research Institute , Yonsei University College of Medicine , Seoul , Korea
| |
Collapse
|
11
|
Loregger A, Raaben M, Tan J, Scheij S, Moeton M, van den Berg M, Gelberg-Etel H, Stickel E, Roitelman J, Brummelkamp T, Zelcer N. Haploid Mammalian Genetic Screen Identifies UBXD8 as a Key Determinant of HMGCR Degradation and Cholesterol Biosynthesis. Arterioscler Thromb Vasc Biol 2017; 37:2064-2074. [PMID: 28882874 PMCID: PMC5671778 DOI: 10.1161/atvbaha.117.310002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 08/29/2017] [Indexed: 01/23/2023]
Abstract
Supplemental Digital Content is available in the text. Objective— The cellular demand for cholesterol requires control of its biosynthesis by the mevalonate pathway. Regulation of HMGCR (3-hydroxy-3-methylglutaryl coenzyme A reductase), a rate-limiting enzyme in this pathway and the target of statins, is a key control point herein. Accordingly, HMGCR is subject to negative and positive regulation. In particular, the ability of oxysterols and intermediates of the mevalonate pathway to stimulate its proteasomal degradation is an exquisite example of metabolically controlled feedback regulation. To define the genetic determinants that govern this process, we conducted an unbiased haploid mammalian genetic screen. Approach and Results— We generated human haploid cells with mNeon fused to endogenous HMGCR using CRISPR/Cas9 and used these cells to interrogate regulation of HMGCR abundance in live cells. This resulted in identification of known and new regulators of HMGCR, and among the latter, UBXD8 (ubiquitin regulatory X domain-containing protein 8), a gene that has not been previously implicated in this process. We demonstrate that UBXD8 is an essential determinant of metabolically stimulated degradation of HMGCR and of cholesterol biosynthesis in multiple cell types. Accordingly, UBXD8 ablation leads to aberrant cholesterol synthesis due to loss of feedback control. Mechanistically, we show that UBXD8 is necessary for sterol-stimulated dislocation of ubiquitylated HMGCR from the endoplasmic reticulum membrane en route to proteasomal degradation, a function dependent on its UBX domain. Conclusions— We establish UBXD8 as a previously unrecognized determinant that couples flux across the mevalonate pathway to control of cholesterol synthesis and demonstrate the feasibility of applying mammalian haploid genetics to study metabolic traits.
Collapse
Affiliation(s)
- Anke Loregger
- From the Department of Medical Biochemistry, Academic Medical Center of the University of Amsterdam, The Netherlands (A.L., J.T., S.S., M.M., M.v.d.B., N.Z.); Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam (M.R., E.S., T.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (T.B.); Cancer GenomiCs.nl, Amsterdam, The Netherlands (T.B.); Bert W. Strassburger Lipid Center, Sheba Medical Center, Tel Hashomer, Israel (H.G.-E., J.R.); and Department of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel (H.G.-E., J.R.)
| | - Matthijs Raaben
- From the Department of Medical Biochemistry, Academic Medical Center of the University of Amsterdam, The Netherlands (A.L., J.T., S.S., M.M., M.v.d.B., N.Z.); Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam (M.R., E.S., T.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (T.B.); Cancer GenomiCs.nl, Amsterdam, The Netherlands (T.B.); Bert W. Strassburger Lipid Center, Sheba Medical Center, Tel Hashomer, Israel (H.G.-E., J.R.); and Department of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel (H.G.-E., J.R.)
| | - Josephine Tan
- From the Department of Medical Biochemistry, Academic Medical Center of the University of Amsterdam, The Netherlands (A.L., J.T., S.S., M.M., M.v.d.B., N.Z.); Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam (M.R., E.S., T.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (T.B.); Cancer GenomiCs.nl, Amsterdam, The Netherlands (T.B.); Bert W. Strassburger Lipid Center, Sheba Medical Center, Tel Hashomer, Israel (H.G.-E., J.R.); and Department of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel (H.G.-E., J.R.)
| | - Saskia Scheij
- From the Department of Medical Biochemistry, Academic Medical Center of the University of Amsterdam, The Netherlands (A.L., J.T., S.S., M.M., M.v.d.B., N.Z.); Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam (M.R., E.S., T.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (T.B.); Cancer GenomiCs.nl, Amsterdam, The Netherlands (T.B.); Bert W. Strassburger Lipid Center, Sheba Medical Center, Tel Hashomer, Israel (H.G.-E., J.R.); and Department of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel (H.G.-E., J.R.)
| | - Martina Moeton
- From the Department of Medical Biochemistry, Academic Medical Center of the University of Amsterdam, The Netherlands (A.L., J.T., S.S., M.M., M.v.d.B., N.Z.); Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam (M.R., E.S., T.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (T.B.); Cancer GenomiCs.nl, Amsterdam, The Netherlands (T.B.); Bert W. Strassburger Lipid Center, Sheba Medical Center, Tel Hashomer, Israel (H.G.-E., J.R.); and Department of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel (H.G.-E., J.R.)
| | - Marlene van den Berg
- From the Department of Medical Biochemistry, Academic Medical Center of the University of Amsterdam, The Netherlands (A.L., J.T., S.S., M.M., M.v.d.B., N.Z.); Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam (M.R., E.S., T.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (T.B.); Cancer GenomiCs.nl, Amsterdam, The Netherlands (T.B.); Bert W. Strassburger Lipid Center, Sheba Medical Center, Tel Hashomer, Israel (H.G.-E., J.R.); and Department of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel (H.G.-E., J.R.)
| | - Hila Gelberg-Etel
- From the Department of Medical Biochemistry, Academic Medical Center of the University of Amsterdam, The Netherlands (A.L., J.T., S.S., M.M., M.v.d.B., N.Z.); Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam (M.R., E.S., T.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (T.B.); Cancer GenomiCs.nl, Amsterdam, The Netherlands (T.B.); Bert W. Strassburger Lipid Center, Sheba Medical Center, Tel Hashomer, Israel (H.G.-E., J.R.); and Department of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel (H.G.-E., J.R.)
| | - Elmer Stickel
- From the Department of Medical Biochemistry, Academic Medical Center of the University of Amsterdam, The Netherlands (A.L., J.T., S.S., M.M., M.v.d.B., N.Z.); Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam (M.R., E.S., T.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (T.B.); Cancer GenomiCs.nl, Amsterdam, The Netherlands (T.B.); Bert W. Strassburger Lipid Center, Sheba Medical Center, Tel Hashomer, Israel (H.G.-E., J.R.); and Department of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel (H.G.-E., J.R.)
| | - Joseph Roitelman
- From the Department of Medical Biochemistry, Academic Medical Center of the University of Amsterdam, The Netherlands (A.L., J.T., S.S., M.M., M.v.d.B., N.Z.); Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam (M.R., E.S., T.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (T.B.); Cancer GenomiCs.nl, Amsterdam, The Netherlands (T.B.); Bert W. Strassburger Lipid Center, Sheba Medical Center, Tel Hashomer, Israel (H.G.-E., J.R.); and Department of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel (H.G.-E., J.R.)
| | - Thijn Brummelkamp
- From the Department of Medical Biochemistry, Academic Medical Center of the University of Amsterdam, The Netherlands (A.L., J.T., S.S., M.M., M.v.d.B., N.Z.); Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam (M.R., E.S., T.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (T.B.); Cancer GenomiCs.nl, Amsterdam, The Netherlands (T.B.); Bert W. Strassburger Lipid Center, Sheba Medical Center, Tel Hashomer, Israel (H.G.-E., J.R.); and Department of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel (H.G.-E., J.R.)
| | - Noam Zelcer
- From the Department of Medical Biochemistry, Academic Medical Center of the University of Amsterdam, The Netherlands (A.L., J.T., S.S., M.M., M.v.d.B., N.Z.); Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam (M.R., E.S., T.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (T.B.); Cancer GenomiCs.nl, Amsterdam, The Netherlands (T.B.); Bert W. Strassburger Lipid Center, Sheba Medical Center, Tel Hashomer, Israel (H.G.-E., J.R.); and Department of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel (H.G.-E., J.R.).
| |
Collapse
|
12
|
Adam AAA, van Wenum M, van der Mark VA, Jongejan A, Moerland PD, Houtkooper RH, Wanders RJA, Oude Elferink RP, Chamuleau RAFM, Hoekstra R. AMC-Bio-Artificial Liver culturing enhances mitochondrial biogenesis in human liver cell lines: The role of oxygen, medium perfusion and 3D configuration. Mitochondrion 2017; 39:30-42. [PMID: 28844938 DOI: 10.1016/j.mito.2017.08.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 08/15/2017] [Accepted: 08/23/2017] [Indexed: 12/18/2022]
Abstract
BACKGROUND Human liver cell lines, like HepaRG and C3A, acquire higher functionality when cultured in the AMC-Bio-Artificial Liver (AMC-BAL). The three main differences between BAL and monolayer culture are the oxygenation (40% vs 20%O2), dynamic vs absent medium perfusion and 3D vs 2D configuration. Here, we investigated the background of the differences between BAL-cultures and monolayers. METHODS We performed whole-genome microarray analysis on HepaRG monolayer and BAL-cultures. Next, mitochondrial biogenesis was studied in monolayer and BAL-cultures of HepaRG and C3A. The driving forces for mitochondrial biogenesis by BAL-culturing were investigated in representative culture models differing in oxygenation level, medium flow or 2D vs 3D configuration. RESULTS Gene-sets related to mitochondrial energy metabolism were most prominently up-regulated in HepaRG-BAL vs monolayer cultures. This was confirmed by a 2.4-fold higher mitochondrial abundance with increased expression of mitochondrial OxPhos complexes. Moreover, the transcript levels of mitochondria-encoded genes were up to 3.6-fold induced and mitochondrial membrane potential activity was 8.3-fold increased in BAL vs monolayers. Culturing with 40% O2, dynamic medium flow and/or in 3D increased the mitochondrial abundance and expression of mitochondrial complexes vs standard monolayer culturing. The stimulatory effect of the BAL culture on mitochondrial biogenesis was confirmed in C3A cells in which mitochondrial abundance increased 2.2-fold with induction of mitochondria-encoded genes. CONCLUSIONS AND GENERAL SIGNIFICANCE The increased functionality of liver cell lines upon AMC-BAL culturing is associated with increased mitochondrial biogenesis. High oxygenation, medium perfusion and 3D configuration contribute to the up-regulation of the mitochondrial biogenesis.
Collapse
Affiliation(s)
- Aziza A A Adam
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (AMC), University of Amsterdam, Postbus 22660, 1100 DD Amsterdam, The Netherlands
| | - Martien van Wenum
- Surgical Laboratory, Academic Medical Center (AMC), University of Amsterdam, Postbus 22660, 1100 DD Amsterdam, The Netherlands
| | - Vincent A van der Mark
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (AMC), University of Amsterdam, Postbus 22660, 1100 DD Amsterdam, The Netherlands; Surgical Laboratory, Academic Medical Center (AMC), University of Amsterdam, Postbus 22660, 1100 DD Amsterdam, The Netherlands
| | - Aldo Jongejan
- Bioinformatics Laboratory, Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam Public Health Research Institute, Academic Medical Center (AMC), University of Amsterdam, Postbus 22660, 1100 DD Amsterdam, The Netherlands
| | - Perry D Moerland
- Bioinformatics Laboratory, Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam Public Health Research Institute, Academic Medical Center (AMC), University of Amsterdam, Postbus 22660, 1100 DD Amsterdam, The Netherlands
| | - Riekelt H Houtkooper
- Laboratory Genetic and Metabolic Diseases, Academic Medical Center (AMC), University of Amsterdam, Postbus 22660, 1100 DD Amsterdam, The Netherlands
| | - Ronald J A Wanders
- Laboratory Genetic and Metabolic Diseases, Academic Medical Center (AMC), University of Amsterdam, Postbus 22660, 1100 DD Amsterdam, The Netherlands
| | - Ronald P Oude Elferink
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (AMC), University of Amsterdam, Postbus 22660, 1100 DD Amsterdam, The Netherlands
| | - Robert A F M Chamuleau
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (AMC), University of Amsterdam, Postbus 22660, 1100 DD Amsterdam, The Netherlands
| | - Ruurdtje Hoekstra
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (AMC), University of Amsterdam, Postbus 22660, 1100 DD Amsterdam, The Netherlands; Surgical Laboratory, Academic Medical Center (AMC), University of Amsterdam, Postbus 22660, 1100 DD Amsterdam, The Netherlands.
| |
Collapse
|
13
|
Deurholt T, ten Bloemendaal L, Chhatta AA, van Wijk ACWA, Weijer K, Seppen J, Elferink RPJO, Chamuleau RAFM, Hoekstra R. In Vitro Functionality of Human Fetal Liver Cells and Clonal Derivatives under Proliferative Conditions. Cell Transplant 2017; 15:811-22. [PMID: 17269451 DOI: 10.3727/000000006783464417] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Mature human hepatocytes are not suitable for large-scale in vitro applications that rely on hepatocyte function, due to their limited availability and insufficient proliferation capacity in vitro. In contrast, human fetal liver cells (HFLC) can be easily expanded in vitro. In this study we evaluated the hepatic function of HFLCs under proliferative conditions, to determine whether HFLCs can replace mature hepatocytes for in vitro applications. HFLCs were isolated from fetal livers of 16 weeks gestation. Hepatic functions of HFLCs were determined in primary culture and after expansion in vitro. Clonal derivatives were selected and tested for hepatic functionality. Results were compared to primary mature human hepatocytes in vitro. No differences were observed between primary HFLCs and mature human hepatocytes in albumin production and mRNA levels of various liver-specific genes. Ureagenesis was 4.4-fold lower and ammonia elimination was absent in HFLCs. Expanding HFLCs decreased hepatic functions and increased cell stretching. In contrast, clonal derivatives had stable functionality and morphology and responded to differentiation stimuli. Although their hepatic functions were higher than in passaged HFLCs, functionality was at least 20 times lower compared to mature human hepatocytes. HFLCs cannot replace mature human hepatocytes in in vitro applications requiring extensive in vitro expansion, because this is associated with decreased hepatic functionality. Selecting functional subpopulations can, at least partly, prevent this. In addition, defining conditions that support hepatic differentiation is necessary to obtain HFLC cultures suitable for in vitro hepatic applications.
Collapse
Affiliation(s)
- Tanja Deurholt
- AMC Liver Center, Academic Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Funkhouser AW, Vahed S, Soriano HE. A “Real Time” PCR Assay to Detect Transplanted Human Liver Cells in RAG-1-/- Mice. Cell Transplant 2017. [DOI: 10.3727/000000001783986990] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Ann W. Funkhouser
- University of Chicago, Department of Pediatrics, University of Chicago Children's Hospital, Chicago, IL 60637
| | - Sabera Vahed
- University of Chicago, Department of Pediatrics, University of Chicago Children's Hospital, Chicago, IL 60637
| | - Humberto E. Soriano
- Northwestern University, Department of Pediatrics, Children's Memorial Hospital, Chicago, IL 60614
| |
Collapse
|
15
|
Hoekstra R, Deurholt T, ten Bloemendaal L, Desille M, van Wijk ACWA, Clement B, Oude Elferink RPJ, van Gulik TM, Chamuleau RAFM. Assessment of in Vitro Applicability of Reversibly Immortalized NKNT-3 Cells and Clonal Derivatives. Cell Transplant 2017; 15:423-433. [DOI: 10.3727/000000006783981873] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
In vitro applications of human hepatocytes, such as bioartificial livers and toxicity assays, require thoroughly testing of human cell lines prior to using them as alternative cell sources. The reversibly immortalized NKNT-3 cell line was reported to show clear in vivo functionality. Here, NKNT-3 cells were tested for their in vitro applicability. Low-passage (P2) and high-passage (P28) NKNT-3 cells and clonal derivatives were characterized for reversion of immortalization, heterogeneity, and hepatic functionality. Reversion with reduced expression of immortalizing agent could be established. However, during culturing the cells lost the capacity to be selected for completed reversion. The phenotypic instability is probably associated with heterogeneity in the culture, as clonal derivatives of P2 cells varied in morphology, growth, and reversion characteristics. The mRNA levels of genes related with hepatic differentiation increased 4–20-fold after reversion. However, the levels never exceeded 0.1% of that detected in liver and no urea production nor ammonia elimination was detected. Additionally, activities of different cytochrome P450s were limited. In conclusion, the NKNT-3 culture is heterogeneous and unstable and the in vitro functionality is relatively low. These findings emphasize that in vivo testing of hepatic cell lines is little informative for predicting their value for in vitro applications.
Collapse
Affiliation(s)
- Ruurdtje Hoekstra
- Surgical Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands
- AMC Liver Center, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Tanja Deurholt
- AMC Liver Center, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Lysbeth ten Bloemendaal
- Surgical Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands
- AMC Liver Center, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Mireille Desille
- INSERM U456, Detoxication and Tissue Repair Unit, University of Rennes I, Rennes, France
| | | | - Bruno Clement
- INSERM U456, Detoxication and Tissue Repair Unit, University of Rennes I, Rennes, France
| | | | - Thomas M. van Gulik
- Surgical Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands
| | | |
Collapse
|
16
|
Bai X, Hong W, Cai P, Chen Y, Xu C, Cao D, Yu W, Zhao Z, Huang M, Jin J. Valproate induced hepatic steatosis by enhanced fatty acid uptake and triglyceride synthesis. Toxicol Appl Pharmacol 2017; 324:12-25. [PMID: 28366540 DOI: 10.1016/j.taap.2017.03.022] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 03/06/2017] [Accepted: 03/28/2017] [Indexed: 02/07/2023]
Abstract
Steatosis is the characteristic type of VPA-induced hepatotoxicity and may result in life-threatening hepatic lesion. Approximately 61% of patients treated with VPA have been diagnosed with hepatic steatosis through ultrasound examination. However, the mechanisms underlying VPA-induced intracellular fat accumulation are not yet fully understood. Here we demonstrated the involvement of fatty acid uptake and lipogenesis in VPA-induced hepatic steatosis in vitro and in vivo by using quantitative real-time PCR (qRT-PCR) analysis, western blotting analysis, fatty acid uptake assays, Nile Red staining assays, and Oil Red O staining assays. Specifically, we found that the expression of cluster of differentiation 36 (CD36), an important fatty acid transport, and diacylglycerol acyltransferase 2 (DGAT2) were significantly up-regulated in HepG2 cells and livers of C57B/6J mice after treatment with VPA. Furthermore, VPA treatment remarkably enhanced the efficiency of fatty acid uptake mediated by CD36, while this effect was abolished by the interference with CD36-specific siRNA. Also, VPA treatment significantly increased DGAT2 expression as a result of the inhibition of mitogen-activated protein kinase kinase (MEK) - extracellular regulated kinase (ERK) pathway; however, DGAT2 knockdown significantly alleviated VPA-induced intracellular lipid accumulation. Additionally, we also found that sterol regulatory element binding protein-1c (SREBP-1c)-mediated fatty acid synthesis may be not involved in VPA-induced hepatic steatosis. Overall, VPA-triggered over-regulation of CD36 and DGAT2 could be helpful for a better understanding of the mechanisms underlying VPA-induced hepatic steatosis and may offer novel therapeutic strategies to combat VPA-induced hepatotoxicity.
Collapse
Affiliation(s)
- Xupeng Bai
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Weipeng Hong
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Peiheng Cai
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yibei Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Chuncao Xu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Di Cao
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Weibang Yu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Zhongxiang Zhao
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Min Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jing Jin
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
17
|
Ginsenoside Rb2 Alleviates Hepatic Lipid Accumulation by Restoring Autophagy via Induction of Sirt1 and Activation of AMPK. Int J Mol Sci 2017; 18:ijms18051063. [PMID: 28534819 PMCID: PMC5454975 DOI: 10.3390/ijms18051063] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/26/2017] [Accepted: 05/09/2017] [Indexed: 12/16/2022] Open
Abstract
Although Panax ginseng is a famous traditional Chinese medicine and has been widely used to treat a variety of metabolic diseases including hyperglycemia, hyperlipidemia, and hepatosteatosis, the effective mediators and molecular mechanisms remain largely unknown. In this study we found that ginsenoside Rb2, one of the major ginsenosides in Panax ginseng, was able to prevent hepatic lipid accumulation through autophagy induction both in vivo and in vitro. Treatment of male db/db mice with Rb2 significantly improved glucose tolerance, decreased hepatic lipid accumulation, and restored hepatic autophagy. In vitro, Rb2 (50 µmol/L) obviously increased autophagic flux in HepG2 cells and primary mouse hepatocytes, and consequently reduced the lipid accumulation induced by oleic acid in combination with high glucose. Western blotting analysis showed that Rb2 partly reversed the high fatty acid in combination with high glucose (OA)-induced repression of autophagic pathways including AMP-activated protein kinase (AMPK) and silent information regulator 1 (sirt1). Furthermore, pharmacological inhibition of the sirt1 or AMPK pathways attenuated these beneficial effects of Rb2 on hepatic autophagy and lipid accumulation. Taken together, these results suggested that Rb2 alleviated hepatic lipid accumulation by restoring autophagy via the induction of sirt1 and activation of AMPK, and resulted in improved nonalcoholic fatty liver disease (NAFLD) and glucose tolerance.
Collapse
|
18
|
Identification of the ER-resident E3 ubiquitin ligase RNF145 as a novel LXR-regulated gene. PLoS One 2017; 12:e0172721. [PMID: 28231341 PMCID: PMC5322959 DOI: 10.1371/journal.pone.0172721] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 02/08/2017] [Indexed: 12/12/2022] Open
Abstract
Cellular cholesterol metabolism is subject to tight regulation to maintain adequate levels of this central lipid molecule. Herein, the sterol-responsive Liver X Receptors (LXRs) play an important role owing to their ability to reduce cellular cholesterol load. In this context, identifying the full set of LXR-regulated genes will contribute to our understanding of their role in cholesterol metabolism. Using global transcriptional analysis we report here the identification of RNF145 as an LXR-regulated target gene. We demonstrate that RNF145 is regulated by LXRs in both human and mouse primary cells and cell lines, and in vivo in mice. Regulation of RNF145 by LXR depends on a functional LXR-element in its proximal promotor. Consistent with LXR-dependent regulation of Rnf145 we show that regulation is lost in macrophages and fibroblasts from Lxrαβ(-/-) mice, and also in vivo in livers of Lxrα(-/-) mice treated with the LXR synthetic ligand T0901317. RNF145 is closely related to RNF139/TRC8, an E3 ligase implicated in control of SREBP processing. However, silencing of RNF145 in HepG2 or HeLa cells does not impair SREBP1/2 processing and sterol-responsive gene expression in these cells. Similar to TRC8, we demonstrate that RNF145 is localized to the ER and that it possesses intrinsic E3 ubiquitin ligase activity. In summary, we report the identification of RNF145 as an ER-resident E3 ubiquitin ligase that is transcriptionally controlled by LXR.
Collapse
|
19
|
Terada M, Horisawa K, Miura S, Takashima Y, Ohkawa Y, Sekiya S, Matsuda-Ito K, Suzuki A. Kupffer cells induce Notch-mediated hepatocyte conversion in a common mouse model of intrahepatic cholangiocarcinoma. Sci Rep 2016; 6:34691. [PMID: 27698452 PMCID: PMC5048166 DOI: 10.1038/srep34691] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 09/19/2016] [Indexed: 12/11/2022] Open
Abstract
Intrahepatic cholangiocarcinoma (ICC) is a malignant epithelial neoplasm composed of cells resembling cholangiocytes that line the intrahepatic bile ducts in portal areas of the hepatic lobule. Although ICC has been defined as a tumor arising from cholangiocyte transformation, recent evidence from genetic lineage-tracing experiments has indicated that hepatocytes can be a cellular origin of ICC by directly changing their fate to that of biliary lineage cells. Notch signaling has been identified as an essential factor for hepatocyte conversion into biliary lineage cells at the onset of ICC. However, the mechanisms underlying Notch signal activation in hepatocytes remain unclear. Here, using a mouse model of ICC, we found that hepatic macrophages called Kupffer cells transiently congregate around the central veins in the liver and express the Notch ligand Jagged-1 coincident with Notch activation in pericentral hepatocytes. Depletion of Kupffer cells prevents the Notch-mediated cell-fate conversion of hepatocytes to biliary lineage cells, inducing hepatocyte apoptosis and increasing mortality in mice. These findings will be useful for uncovering the pathogenic mechanism of ICC and developing prevenient and therapeutic strategies for this refractory disease.
Collapse
Affiliation(s)
- Maiko Terada
- Division of Organogenesis and Regeneration, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Kenichi Horisawa
- Division of Organogenesis and Regeneration, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Shizuka Miura
- Division of Organogenesis and Regeneration, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yasuo Takashima
- Division of Organogenesis and Regeneration, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.,Core Research for Evolutional Science and Technology, The Japan Agency for Medical Research and Development, 1-7-1 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Sayaka Sekiya
- Division of Organogenesis and Regeneration, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Kanae Matsuda-Ito
- Division of Organogenesis and Regeneration, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Atsushi Suzuki
- Division of Organogenesis and Regeneration, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.,Core Research for Evolutional Science and Technology, The Japan Agency for Medical Research and Development, 1-7-1 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| |
Collapse
|
20
|
Takashima Y, Terada M, Udono M, Miura S, Yamamoto J, Suzuki A. Suppression of lethal-7b and miR-125a/b Maturation by Lin28b Enables Maintenance of Stem Cell Properties in Hepatoblasts. Hepatology 2016; 64:245-60. [PMID: 26990797 DOI: 10.1002/hep.28548] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 03/06/2016] [Indexed: 12/28/2022]
Abstract
UNLABELLED In liver development, hepatoblasts that act as hepatic stem/progenitor cells proliferate and differentiate into both hepatocytes and cholangiocytes to form liver tissues. Although numerous factors contribute to this event, little is known about the roles of microRNAs in hepatoblast proliferation and differentiation. In this study, we focused on the lineage-28 (Lin28) family proteins, which are required for microRNA regulation in pluripotent stem cells and cancer cells, and investigated their roles as regulatory factors for the properties of hepatoblasts. CONCLUSION Lin28b was specifically expressed in hepatoblasts, and its suppression induced growth arrest and cholangiocyte differentiation of hepatoblasts; mechanistically, Lin28b positively regulates the expression of Lin28b itself and cell cycle-related proteins in hepatoblasts by suppressing the maturation of target microRNAs, lethal-7b and miR-125a/b, enabling maintenance of the stem cell properties of hepatoblasts, such as their capabilities for proliferation and bi-lineage differentiation, during liver development. (Hepatology 2016;64:245-260).
Collapse
Affiliation(s)
- Yasuo Takashima
- Division of Organogenesis and Regeneration, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Maiko Terada
- Division of Organogenesis and Regeneration, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Miyako Udono
- Division of Organogenesis and Regeneration, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Shizuka Miura
- Division of Organogenesis and Regeneration, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Junpei Yamamoto
- Division of Organogenesis and Regeneration, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Atsushi Suzuki
- Division of Organogenesis and Regeneration, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.,Core Research for Evolutional Science and Technology, The Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, Japan
| |
Collapse
|
21
|
Yanagida A, Mizuno N, Yamazaki Y, Kato-Itoh M, Umino A, Sato H, Ito K, Yamaguchi T, Nakauchi H, Kamiya A. Investigation of bipotent differentiation of hepatoblasts using inducible diphtheria toxin receptor-transgenic mice. Hepatol Res 2016; 46:816-28. [PMID: 26584962 DOI: 10.1111/hepr.12622] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 11/05/2015] [Accepted: 11/05/2015] [Indexed: 02/08/2023]
Abstract
AIM Hepatic progenitor cells, called hepatoblasts, are highly proliferative and exhibit bipotential differentiation into hepatocytes and cholangiocytes in the fetal liver. Thus, they are the ideal source for transplantation therapy. Although several studies have been performed in vitro, the molecular mechanisms regulating hepatoblast differentiation in vivo following transplantation remain poorly understood. The aim of this study was to investigate an in vivo model to analyze hepatoblast bipotency and proliferative ability. METHODS Hepatic transplantation model using Cre-inducible diphtheria toxin receptor-transgenic mice (iDTR), and albafpCre mice expressing Cre under the control of albumin and α-fetoprotein (AFP) regulatory elements were established. Fresh hepatoblasts were transplanted into diphtheria toxin (DT)-injected iDTRalbafpCre mice and we analyzed their differentiation and proliferation abilities by immunostaining and gene expression profiles. RESULTS Fresh hepatoblasts transplanted into DT-injected iDTRalbafpCre mice engrafted and differentiated into both hepatocytes and cholangiocytes. Additionally, the number of engrafted hepatoblast-derived hepatocytes increased following partial hepatectomy and serial DT injections. Expression levels of hepatic functional genes in transplanted hepatoblast-derived hepatocytes were similar to that of normal hepatocytes. CONCLUSION In our iDTRalbafpCre transplantation model, fresh hepatoblasts could differentiate into hepatocytes and cholangiocytes. In addition, these donor cells were induced to proliferate by the following liver injury stimulation. This result suggests that this model is valuable for investigating hepatoblast differentiation pathways in vivo.
Collapse
Affiliation(s)
- Ayaka Yanagida
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan.,Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Naoak Mizuno
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Yuji Yamazaki
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Megumi Kato-Itoh
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Ayumi Umino
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Hideyuki Sato
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Keiichi Ito
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan.,Laboratory of Biochemistry and Molecular Biology, The Rockefeller University, New York, USA
| | - Tomoyuki Yamaguchi
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Hiromitsu Nakauchi
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, USA
| | - Akihide Kamiya
- Department of Molecular Life Sciences, Tokai University School of Medicine, Isehara, Japan
| |
Collapse
|
22
|
Song YM, Lee YH, Kim JW, Ham DS, Kang ES, Cha BS, Lee HC, Lee BW. Metformin alleviates hepatosteatosis by restoring SIRT1-mediated autophagy induction via an AMP-activated protein kinase-independent pathway. Autophagy 2015; 11:46-59. [PMID: 25484077 DOI: 10.4161/15548627.2014.984271] [Citation(s) in RCA: 230] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Metformin activates both PRKA and SIRT1. Furthermore, autophagy is induced by either the PRKA-MTOR-ULK1 or SIRT1-FOXO signaling pathways. We aimed to elucidate the mechanism by which metformin alleviates hepatosteatosis by examining the molecular interplay between SIRT1, PRKA, and autophagy. ob/ob mice were divided into 3 groups: one with ad libitum feeding of a standard chow diet, one with 300 mg/kg intraperitoneal metformin injections, and one with 3 g/d caloric restriction (CR) for a period of 4 wk. Primary hepatocytes or HepG2 cells were treated with oleic acid (OA) plus high glucose in the absence or presence of metformin. Both CR and metformin significantly improved body weight and glucose homeostasis, along with hepatic steatosis, in ob/ob mice. Furthermore, CR and metformin both upregulated SIRT1 expression and also stimulated autophagy induction and flux in vivo. Metformin also prevented OA with high glucose-induced suppression of both SIRT1 expression and SIRT1-dependent activation of autophagy machinery, thereby alleviating intracellular lipid accumulation in vitro. Interestingly, metformin treatment upregulated SIRT1 expression and activated PRKA even after siRNA-mediated knockdown of PRKAA1/2 and SIRT1, respectively. Taken together, these results suggest that metformin alleviates hepatic steatosis through PRKA-independent, SIRT1-mediated effects on the autophagy machinery.
Collapse
Key Words
- 3MA, 3-methyladenine
- CQ, chloroquine
- CR, caloric restriction
- GOT1/AST, glutamic-oxaloacetic transaminase 1, soluble
- GPT/ALT, glutamic-pyruvate transaminase (alanine aminotransferase)
- IPGTTs, intraperitoneal glucose tolerance tests
- MTOR, mechanistic target of rapamycin
- Met, metformin
- NAFLD, nonalcoholic fatty liver disease
- OA, oleic acid
- ORO, Oil Red O
- PRKA
- PRKA, protein kinase, AMP-activated
- SIRT1
- SIRT1, sirtuin 1
- T-CHO, total cholesterol
- TG, triglyceride
- autophagy
- hepatoseatosis
- metformin
- siRNA, short interfering RNA
Collapse
Affiliation(s)
- Young Mi Song
- a Brain Korea 21 PLUS Project for Medical Science ; Yonsei University College of Medicine ; Seoul , Korea
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Exogenous administration of DLK1 ameliorates hepatic steatosis and regulates gluconeogenesis via activation of AMPK. Int J Obes (Lond) 2015; 40:356-65. [PMID: 26315841 DOI: 10.1038/ijo.2015.173] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 08/07/2015] [Accepted: 08/13/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND/OBJECTIVES Activation of Notch signaling pathologically enhances lipogenesis and gluconeogenesis in the liver causing non-alcoholic fatty liver disease (NAFLD) and diabetes. Delta-like 1 homolog (DLK1), an imprinted gene that can modulate adipogenesis and muscle development in mice, was found as an inhibitory regulator of Notch signaling. Therefore, we investigated the metabolic effect of exogenous DLK1 in vitro and in vivo. SUBJECTS/METHODS A soluble DLK1 peptide was generated with fusion between a human Fc fragment and extracellular domain of DLK1. Male db/db mice were randomly assigned to two groups: vehicle treated and DLK1-treated group (25 mg kg(-1), intraperitoneal injection, twice a week for 4 weeks). Primary mice hepatocytes and HepG2 cells were used for in vitro experiments. RESULTS After 4 weeks of DLK1 administration, hepatic triglyceride content and lipid droplets in liver tissues, as well as serum levels of liver enzymes, were markedly decreased in db/db mice. DLK1 treatment induced phosphorylation of AMPK and ACC and suppressed nuclear expression of SREBP-1c in the mouse liver or hepatocytes, indicating regulation of fatty acid oxidation and synthesis pathways. Furthermore, DLK1-treated mice showed significantly lower levels of fasting and random glucose, with improved glucose and insulin tolerance compared with the vehicle-treated group. Macrophage infiltration and proinflammatory cytokine levels in the epididymal fat were decreased in DLK1-treated db/db mice. Moreover, DLK1 suppressed glucose production from hepatocytes, which was blocked after co-administration of an AMPK inhibitor, compound C. DLK1-treated hepatocytes and mouse liver tissues showed lower PEPCK and G6Pase expression. DLK1 triggered AKT phosphorylation followed by cytosolic translocation of FOXO1 from the nucleus in hepatocytes. CONCLUSIONS The present study demonstrated that exogenous administration of DLK1 reduced hepatic steatosis and hyperglycemia via AMPK activation in the liver. This result suggests that DLK1 may be a novel therapeutic approach for treating NAFLD and diabetes.
Collapse
|
24
|
Yanagida A, Chikada H, Ito K, Umino A, Kato-Itoh M, Yamazaki Y, Sato H, Kobayashi T, Yamaguchi T, Nakayama KI, Nakauchi H, Kamiya A. Liver maturation deficiency in p57(Kip2)-/- mice occurs in a hepatocytic p57(Kip2) expression-independent manner. Dev Biol 2015; 407:331-43. [PMID: 26165599 DOI: 10.1016/j.ydbio.2015.07.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 07/02/2015] [Accepted: 07/06/2015] [Indexed: 12/29/2022]
Abstract
Fetal hepatic stem/progenitor cells, hepatoblasts, are highly proliferative cells and the source of both hepatocytes and cholangiocytes. In contrast, mature hepatocytes have a low proliferative potency and high metabolic functions. Cell proliferation is regulated by cell cycle-related molecules. However, the correlation between cell cycle regulation and hepatic maturation are still unknown. To address this issue, we revealed that the cell cycle inhibitor p57(Kip2) was expressed in the hepatoblasts and mesenchymal cells of fetal liver in a spatiotemporal manner. In addition, we found that hepatoblasts in p57(Kip2)-/- mice were highly proliferative and had deficient maturation compared with those in wild-type (WT) mice. However, there were no remarkable differences in the expression levels of cell cycle- and bipotency-related genes except for Ccnd2. Furthermore, p57(Kip2)-/- hepatoblasts could differentiate into mature hepatocytes in p57(Kip2)-/- and WT chimeric mice, suggesting that the intrinsic activity of p57(Kip2) does not simply regulate hepatoblast maturation.
Collapse
Affiliation(s)
- Ayaka Yanagida
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Hiromi Chikada
- Department of Molecular Life Sciences, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa 259-1193, Japan
| | - Keiichi Ito
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Ayumi Umino
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Megumi Kato-Itoh
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Yuji Yamazaki
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Hideyuki Sato
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Toshihiro Kobayashi
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Tomoyuki Yamaguchi
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Keiichi I Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Hiromitsu Nakauchi
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305-5461, USA
| | - Akihide Kamiya
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan; Department of Molecular Life Sciences, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa 259-1193, Japan.
| |
Collapse
|
25
|
van Dijk R, Kremer AE, Smit W, van den Elzen B, van Gulik T, Gouma D, Lameris JS, Bikker H, Enemuo V, Stokkers PCF, Feist M, Bosma P, Jansen PLM, Beuers U. Characterization and treatment of persistent hepatocellular secretory failure. Liver Int 2015; 35:1478-88. [PMID: 24905729 DOI: 10.1111/liv.12603] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 05/18/2014] [Indexed: 01/04/2023]
Abstract
BACKGROUND & AIMS Hepatocellular secretory failure induced by drugs, toxins or transient biliary obstruction may sometimes persist for months after removal of the initiating factor and may then be fatal without liver transplantation. We characterized patients with severe persistent hepatocellular secretory failure (PHSF) and treated them with the pregnane X receptor (PXR) agonist, rifampicin. We also studied the effect of rifampicin on PXR-dependent expression of genes involved in biotransformation and secretion in vitro. METHODS Thirteen patients (age 18-81 years, 6 male) with hepatocellular secretory failure that persisted after removal of the inducing factor (drugs/toxin: 9) or biliary obstruction (4) were identified over 6 years. Six of these patients were screened for ATP8B1 or ABCB11 mutations. All were treated with rifampicin (300 mg daily) for 1-10 weeks. Expression of genes involved in biotransformation and secretion was determined by rtPCR in human hepatocytes and intestinal cells incubated with rifampicin (10 μmol/L). RESULTS Serum bilirubin of patients with PHSF ranged from 264 to 755 μmol/L. Normal γGT was found in 10/13 patients of whom 3/6 tested positive for ATP8B1/ABCB11 mutations. Serum bilirubin declined to <33 μmol/L after 1-10 weeks of rifampicin treatment. In vitro, rifampicin PXR-dependently upregulated biotransformation phase 1 (CYP3A4), phase 2 (UGT1A1) and phase 3 (MRP2) enzymes/carriers as well as the basolateral bile salt exporter OSTβ. CONCLUSION Persistent hepatocellular secretory failure may develop in carriers of transporter gene mutations. In severe cases, rifampicin may represent an effective therapeutic option of PHSF. PXR-dependent induction of CYP3A4, UGT1A1, MRP2 and OSTβ could contribute to the anticholestatic effect of rifampicin in PHSF.
Collapse
Affiliation(s)
- Remco van Dijk
- Department of Gastroenterology & Hepatology and Tytgat Institute for Liver and Intestinal Research, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Miura S, Suzuki A. Acquisition of lipid metabolic capability in hepatocyte-like cells directly induced from mouse fibroblasts. Front Cell Dev Biol 2014; 2:43. [PMID: 25364750 PMCID: PMC4207052 DOI: 10.3389/fcell.2014.00043] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 08/05/2014] [Indexed: 01/15/2023] Open
Abstract
Recently, the numbers of patients with non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) have increased worldwide. NAFLD and NASH are known as risk factors for liver cirrhosis and hepatocellular carcinoma. Because many factors can promote the progression of NAFLD and NASH, the treatment of these patients involves various strategies. Thus, it is desired that drugs for patients with NAFLD and NASH should be developed more easily and rapidly using cultures of primary hepatocytes. However, it is difficult to use hepatocytes as a tool for drug screening, because these cells cannot be functionally maintained in culture. Thus, in this study, we sought to examine whether induced hepatocyte-like (iHep) cells, which were directly induced from mouse dermal fibroblasts by infection with a retrovirus expressing Hnf4α and Foxa3, possess the potential for lipid metabolism, similar to hepatocytes. Our data showed that iHep cells were capable of synthesizing lipids from a cis-unsaturated fatty acid, a trans-unsaturated fatty acid, and a saturated fatty acid, accumulating the synthesized lipids in cellular vesicles, and secreting the lipids into the culture medium. Moreover, the lipid synthesis in iHep cells was significantly inhibited in cultures with lipid metabolism improvers. These results demonstrate that iHep cells could be useful not only for screening of drugs for patients with NAFLD and NASH, but also for elucidation of the mechanisms underlying hereditary lipid metabolism disorders, as an alternative to hepatocytes.
Collapse
Affiliation(s)
- Shizuka Miura
- Division of Organogenesis and Regeneration, Medical Institute of Bioregulation, Kyushu University Fukuoka, Japan
| | - Atsushi Suzuki
- Division of Organogenesis and Regeneration, Medical Institute of Bioregulation, Kyushu University Fukuoka, Japan ; Core Research for Evolutional Science and Technology, Japan Science and Technology Agency Saitama, Japan
| |
Collapse
|
27
|
Ito K, Yamazaki S, Yamamoto R, Tajima Y, Yanagida A, Kobayashi T, Kato-Itoh M, Kakuta S, Iwakura Y, Nakauchi H, Kamiya A. Gene targeting study reveals unexpected expression of brain-expressed X-linked 2 in endocrine and tissue stem/progenitor cells in mice. J Biol Chem 2014; 289:29892-911. [PMID: 25143383 DOI: 10.1074/jbc.m114.580084] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Identification of genes specifically expressed in stem/progenitor cells is an important issue in developmental and stem cell biology. Genome-wide gene expression analyses in liver cells performed in this study have revealed a strong expression of X-linked genes that include members of the brain-expressed X-linked (Bex) gene family in stem/progenitor cells. Bex family genes are expressed abundantly in the neural cells and have been suggested to play important roles in the development of nervous tissues. However, the physiological role of its individual members and the precise expression pattern outside the nervous system remain largely unknown. Here, we focused on Bex2 and examined its role and expression pattern by generating knock-in mice; the enhanced green fluorescence protein (EGFP) was inserted into the Bex2 locus. Bex2-deficient mice were viable and fertile under laboratory growth conditions showing no obvious phenotypic abnormalities. Through an immunohistochemical analysis and flow cytometry-based approach, we observed unique EGFP reporter expression patterns in endocrine and stem/progenitor cells of the liver, pyloric stomach, and hematopoietic system. Although Bex2 seems to play redundant roles in vivo, these results suggest the significance and potential applications of Bex2 in studies of endocrine and stem/progenitor cells.
Collapse
Affiliation(s)
- Keiichi Ito
- From the Division of Stem Cell Therapy, Center for Stem Cell and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
| | - Satoshi Yamazaki
- From the Division of Stem Cell Therapy, Center for Stem Cell and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
| | - Ryo Yamamoto
- From the Division of Stem Cell Therapy, Center for Stem Cell and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo 108-8639, Japan, the Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California 94305, and
| | - Yoko Tajima
- From the Division of Stem Cell Therapy, Center for Stem Cell and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
| | - Ayaka Yanagida
- From the Division of Stem Cell Therapy, Center for Stem Cell and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
| | - Toshihiro Kobayashi
- the NAKAUCHI Stem Cell and Organ Regeneration Project, Japan Science and Technology Agency, Chiyoda-ku, Tokyo 102-8666, Japan, the Wellcome Trust Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, United Kingdom
| | - Megumi Kato-Itoh
- From the Division of Stem Cell Therapy, Center for Stem Cell and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo 108-8639, Japan, the NAKAUCHI Stem Cell and Organ Regeneration Project, Japan Science and Technology Agency, Chiyoda-ku, Tokyo 102-8666, Japan
| | - Shigeru Kakuta
- the Department of Biomedical Science, Graduate School of Agriculture and Life Sciences, University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Yoichiro Iwakura
- the Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, 2669 Yamazaki, Noda, Chiba 278-0022, Japan
| | - Hiromitsu Nakauchi
- From the Division of Stem Cell Therapy, Center for Stem Cell and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo 108-8639, Japan, the Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California 94305, and the NAKAUCHI Stem Cell and Organ Regeneration Project, Japan Science and Technology Agency, Chiyoda-ku, Tokyo 102-8666, Japan
| | - Akihide Kamiya
- From the Division of Stem Cell Therapy, Center for Stem Cell and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo 108-8639, Japan, the Laboratory of Stem Cell Therapy, Institute of Innovative Science and Technology, Tokai University, 143 Shimokasuya, Isehara, Kanagawa 259-1143, Japan
| |
Collapse
|
28
|
Tiruvannamalai-Annamalai R, Armant DR, Matthew HWT. A glycosaminoglycan based, modular tissue scaffold system for rapid assembly of perfusable, high cell density, engineered tissues. PLoS One 2014; 9:e84287. [PMID: 24465401 PMCID: PMC3896358 DOI: 10.1371/journal.pone.0084287] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 11/20/2013] [Indexed: 12/16/2022] Open
Abstract
The limited ability to vascularize and perfuse thick, cell-laden tissue constructs has hindered efforts to engineer complex tissues and organs, including liver, heart and kidney. The emerging field of modular tissue engineering aims to address this limitation by fabricating constructs from the bottom up, with the objective of recreating native tissue architecture and promoting extensive vascularization. In this paper, we report the elements of a simple yet efficient method for fabricating vascularized tissue constructs by fusing biodegradable microcapsules with tunable interior environments. Parenchymal cells of various types, (i.e. trophoblasts, vascular smooth muscle cells, hepatocytes) were suspended in glycosaminoglycan (GAG) solutions (4%/1.5% chondroitin sulfate/carboxymethyl cellulose, or 1.5 wt% hyaluronan) and encapsulated by forming chitosan-GAG polyelectrolyte complex membranes around droplets of the cell suspension. The interior capsule environment could be further tuned by blending collagen with or suspending microcarriers in the GAG solution These capsule modules were seeded externally with vascular endothelial cells (VEC), and subsequently fused into tissue constructs possessing VEC-lined, inter-capsule channels. The microcapsules supported high density growth achieving clinically significant cell densities. Fusion of the endothelialized, capsules generated three dimensional constructs with an embedded network of interconnected channels that enabled long-term perfusion culture of the construct. A prototype, engineered liver tissue, formed by fusion of hepatocyte-containing capsules exhibited urea synthesis rates and albumin synthesis rates comparable to standard collagen sandwich hepatocyte cultures. The capsule based, modular approach described here has the potential to allow rapid assembly of tissue constructs with clinically significant cell densities, uniform cell distribution, and endothelialized, perfusable channels.
Collapse
Affiliation(s)
| | - David Randall Armant
- Departments of Obstetrics & Gynecology, Wayne State University, Detroit, Michigan, United States of America
- Program in Reproductive & Adult Endocrinology, National Institute of Child Health & Human Development, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Howard W. T. Matthew
- Department of Biomedical Engineering, Wayne State University, Detroit, Michigan, United States of America
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, Michigan, United States of America
| |
Collapse
|
29
|
Brugman MH, Suerth JD, Rothe M, Suerbaum S, Schambach A, Modlich U, Kustikova O, Baum C. Evaluating a ligation-mediated PCR and pyrosequencing method for the detection of clonal contribution in polyclonal retrovirally transduced samples. Hum Gene Ther Methods 2013; 24:68-79. [PMID: 23384086 DOI: 10.1089/hgtb.2012.175] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Retroviral gene transfer has proven therapeutic potential in clinical gene therapy trials but may also cause abnormal cell growth via perturbation of gene expression in the locus surrounding the insertion site. By establishing clonal marks, retroviral insertions are also used to describe the regenerative potential of individual cells. Deep sequencing approaches have become the method of choice to study insertion profiles in preclinical models and clinical trials. We used a protocol combining ligation-mediated polymerase chain reaction (LM-PCR) and pyrosequencing for insertion profiling and quantification in cells of various tissues transduced with various retroviral vectors. The presented method allows simultaneous analysis of a multitude of DNA-barcoded samples per pyrosequencing run, thereby allowing cost-effective insertion screening in studies with multiple samples. In addition, we investigated whether the number of pyrosequencing reads can be used to quantify clonal abundance. By comparing pyrosequencing reads against site-specific quantitative PCR and by performing spike-in experiments, we show that considerable variation exists in the quantification of insertion sites even when present in the same clone. Our results suggest that the protocol used here and similar approaches might misinterpret abundance clones defined by insertion sites, unless careful calibration measures are taken. The crucial variables causing this variation need to be defined and methodological improvements are required to establish pyrosequencing reads as a quantification measure in polyclonal situations.
Collapse
Affiliation(s)
- Martijn H Brugman
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Dash A, Simmers MB, Deering TG, Berry DJ, Feaver RE, Hastings NE, Pruett TL, LeCluyse EL, Blackman BR, Wamhoff BR. Hemodynamic flow improves rat hepatocyte morphology, function, and metabolic activity in vitro. Am J Physiol Cell Physiol 2013; 304:C1053-63. [PMID: 23485712 DOI: 10.1152/ajpcell.00331.2012] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
In vitro primary hepatocyte systems typically elicit drug induction and toxicity responses at concentrations much higher than corresponding in vivo or clinical plasma C(max) levels, contributing to poor in vitro-in vivo correlations. This may be partly due to the absence of physiological parameters that maintain metabolic phenotype in vivo. We hypothesized that restoring hemodynamics and media transport would improve hepatocyte architecture and metabolic function in vitro compared with nonflow cultures. Rat hepatocytes were cultured for 2 wk either in nonflow collagen gel sandwiches with 48-h media changes or under controlled hemodynamics mimicking sinusoidal circulation within a perfused Transwell device. Phenotypic, functional, and metabolic parameters were assessed at multiple times. Hepatocytes in the devices exhibited polarized morphology, retention of differentiation markers [E-cadherin and hepatocyte nuclear factor-4α (HNF-4α)], the canalicular transporter [multidrug-resistant protein-2 (Mrp-2)], and significantly higher levels of liver function compared with nonflow cultures over 2 wk (albumin ~4-fold and urea ~5-fold). Gene expression of cytochrome P450 (CYP) enzymes was significantly higher (fold increase over nonflow: CYP1A1: 53.5 ± 10.3; CYP1A2: 64.0 ± 15.1; CYP2B1: 15.2 ± 2.9; CYP2B2: 2.7 ± 0.8; CYP3A2: 4.0 ± 1.4) and translated to significantly higher basal enzyme activity (device vs. nonflow: CYP1A: 6.26 ± 2.41 vs. 0.42 ± 0.015; CYP1B: 3.47 ± 1.66 vs. 0.4 ± 0.09; CYP3A: 11.65 ± 4.70 vs. 2.43 ± 0.56) while retaining inducibility by 3-methylcholanthrene and dexamethasone (fold increase over DMSO: CYP1A = 27.33 and CYP3A = 4.94). These responses were observed at concentrations closer to plasma levels documented in vivo in rats. The retention of in vivo-like hepatocyte phenotype and metabolic function coupled with drug response at more physiological concentrations emphasizes the importance of restoring in vivo physiological transport parameters in vitro.
Collapse
Affiliation(s)
- A Dash
- HemoShear, LLC, Charlottesville, VA 22902, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Jie Q, Tang Y, Deng Y, Li Y, Shi Y, Gao C, Xing M, Wang D, Liu L, Yao P. Bilirubin participates in protecting of heme oxygenase-1 induction by quercetin against ethanol hepatotoxicity in cultured rat hepatocytes. Alcohol 2013; 47:141-8. [PMID: 23265624 DOI: 10.1016/j.alcohol.2012.10.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Revised: 10/12/2012] [Accepted: 10/19/2012] [Indexed: 12/17/2022]
Abstract
To attenuate alcohol liver disease (ALD) is extremely urgent since ALD has been emerged as a major liver disease. The aim of the present study is to investigate the hepatoprotective effect against ethanol-induced injury of bilirubin, a product of heme metabolism degradation via HO and biliverdin reductase catalysis. Ethanol-incubated primary rat hepatocytes (100 mmol/L) were treated by quercetin, bilirubin, inflammatory factors, and/or HO-1 inducer/inhibitor for 24 h, and the cellular damage was assayed. Quercetin lowered ethanol-induced glutathione depletion and superoxide dismutase inactivation, inhibited the overproduction of malondialdehyde and reactive oxygen species, and decreased the leakage of cellular aspartate aminotransferase and lactate dehydrogenase, accompanying the normalization of bilirubin level. The effect of quercetin was mimicked by exogenous bilirubin in a dose-dependent manner to some extent (within 25 μmol/L) and pharmacological HO-1 inducer hemin, but abolished by HO-1 inhibitor zinc protoporphyrin-IX. Inflammatory challenge of TNF-α plus IL-6 further aggravated ethanol-induced oxidative damage, which was also attenuated by bilirubin in part. These findings shed a light on the anti-oxidative and anti-inflammatory role of bilirubin released from quercetin/HO-1 and biliverdin reductase pathway against ethanol hepatotoxicity and highlight a prospective strategy of nutritional intervention for ALD by naturally occurring quercetin to induce HO-1 with the release of bioactive end-products.
Collapse
|
32
|
Patel JP, Hamdy DA, El-kadi AO, Brocks DR. Effect of serum lipoproteins on stereoselective halofantrine metabolism by rat hepatocytes. Chirality 2012; 24:558-65. [DOI: 10.1002/chir.22054] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Accepted: 03/19/2012] [Indexed: 11/06/2022]
Affiliation(s)
- Jigar P. Patel
- Faculty of Pharmacy; University of Montreal; Montreal Quebec Canada
| | | | - A. O. El-kadi
- Faculty of Pharmacy and Pharmaceutical Sciences; University of Alberta; Edmonton Alberta Canada
| | - Dion R. Brocks
- Faculty of Pharmacy and Pharmaceutical Sciences; University of Alberta; Edmonton Alberta Canada
| |
Collapse
|
33
|
Ito H, Kamiya A, Ito K, Yanagida A, Okada K, Nakauchi H. In vitro expansion and functional recovery of mature hepatocytes from mouse adult liver. Liver Int 2012; 32:592-601. [PMID: 22222094 DOI: 10.1111/j.1478-3231.2011.02741.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Accepted: 12/08/2011] [Indexed: 02/13/2023]
Abstract
BACKGROUND Mature hepatocytes retain the ability to regenerate the liver lobule fully in vivo following injury. Several cytokines and soluble factors (hepatocyte growth factors, epidermal growth factors, insulin and nicotinamide) are known to be important for proliferation of mature hepatocytes in vitro. However, hepatocytes monolayer-cultured on extracellular matrices have gradually lost their specific functions, particularly those in drug metabolism. AIM We have explored and established a new culture system for expansion of functional hepatocytes. METHODS We evaluated two approaches for efficient expansion of mature hepatocytes: (i) Co-culture with mouse embryonic fibroblasts (MEF); (ii) Addition to culture of inhibitors of cell signals involved in liver regeneration. After expansion steps, 3-dimensional spheroid-forming culture was used to re-induce mature hepatocellular function. RESULTS The addition of inhibitors for tumour growth factor (TGF) β and glycogen synthase kinase (GSK) 3β efficiently induced in vitro expansion of mature hepatocytes. Although expression of hepatocellular functional genes decreased after expansion in monolayer culture, their expression and the activity of cytochrome P450 enzymes significantly increased with spheroid formation. Furthermore, when hepatocytes were co-cultured with MEF, addition of a MAPK/ERK kinase (MEK) inhibitor at the spheroid formation step enhanced drug-metabolism-related gene expression. CONCLUSION Combination of the MEF co-culture system with the addition of inhibitors of TGFβ and GSK3β induced in vitro expansion of hepatocytes. Moreover, expression of mature hepatocellular genes and the activity of drug-metabolism enzymes in expanded hepatocytes were re-induced after spheroid culture.
Collapse
Affiliation(s)
- Hidenori Ito
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
34
|
Straub BK, Rickelt S, Zimbelmann R, Grund C, Kuhn C, Iken M, Ott M, Schirmacher P, Franke WW. E-N-cadherin heterodimers define novel adherens junctions connecting endoderm-derived cells. ACTA ACUST UNITED AC 2011; 195:873-87. [PMID: 22105347 PMCID: PMC3257573 DOI: 10.1083/jcb.201106023] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Intercellular junctions play a pivotal role in tissue development and function and also in tumorigenesis. In epithelial cells, decrease or loss of E-cadherin, the hallmark molecule of adherens junctions (AJs), and increase of N-cadherin are widely thought to promote carcinoma progression and metastasis. In this paper, we show that this "cadherin switch" hypothesis does not hold for diverse endoderm-derived cells and cells of tumors derived from them. We show that the cadherins in a major portion of AJs in these cells can be chemically cross-linked in E-N heterodimers. We also show that cells possessing E-N heterodimer AJs can form semistable hemihomotypic AJs with purely N-cadherin-based AJs of mesenchymally derived cells, including stroma cells. We conclude that these heterodimers are the major AJ constituents of several endoderm-derived tissues and tumors and that the prevailing concept of antagonistic roles of these two cadherins in developmental and tumor biology has to be reconsidered.
Collapse
Affiliation(s)
- Beate K Straub
- Helmholtz Group for Cell Biology, German Cancer Research Center, 69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
|
36
|
Epidermal growth factor improves lentivirus vector gene transfer into primary mouse hepatocytes. Gene Ther 2011; 19:425-34. [PMID: 21850050 DOI: 10.1038/gt.2011.117] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Partial resistance of primary mouse hepatocytes to lentiviral (LV) vector transduction poses a challenge for ex vivo gene therapy protocols in models of monogenetic liver disease. We thus sought to optimize ex vivo LV gene transfer while preserving the hepatocyte integrity for subsequent transplantation into recipient animals. We found that culture media supplemented with epidermal growth factor (EGF) and, to a lesser extent, hepatocyte growth factor (HGF) markedly improved transduction efficacy at various multiplicities of infection. Up to 87% of primary hepatocytes were transduced in the presence of 10 ng EGF, compared with ~30% in standard culture medium (SCMs). The increased number of transgene-expressing cells correlated with increased nuclear import and more integrated pro-viral copies per cell. Higher LV transduction efficacy was not associated with proliferation, as transduction capacity of gammaretroviral vectors remained low (<1%). Finally, we developed an LV transduction protocol for short-term (maximum 24 h) adherent hepatocyte cultures. LV-transduced hepatocytes showed liver repopulation capacities similar to freshly isolated hepatocytes in alb-uPA mouse recipients. Our findings highlight the importance of EGF for efficient LV transduction of primary hepatocytes in culture and should facilitate studies of LV gene transfer in mouse models of monogenetic liver disease.
Collapse
|
37
|
Raj VP, Chandrasekhar RH, P V, S A D, Rao MC, Rao VJ, Nitesh K. In vitro and in vivo hepatoprotective effects of the total alkaloid fraction of Hygrophila auriculata leaves. Indian J Pharmacol 2011; 42:99-104. [PMID: 20711375 PMCID: PMC2907024 DOI: 10.4103/0253-7613.64500] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2008] [Revised: 10/12/2009] [Accepted: 04/30/2010] [Indexed: 11/04/2022] Open
Abstract
OBJECTIVE To investigate the total alkaloid fraction of the methanol extract of leaves of Hygrophila auriculata for its hepatoprotective activity against CCl4-induced toxicity in freshly isolated rat hepatocytes, HepG2 cells, and animal models. MATERIALS AND METHODS Mature leaves of H. auriculata were collected, authenticated, and subjected to methanolic extraction followed by isolation of total alkaloid fraction. Freshly isolated rat hepatocytes were exposed to CCl4 (1%) along with/without various concentrations of the total alkaloid fraction (80-40 microg/ml). Protection of human liver-derived HepG2 cells against CCl4-induced damage was determined by the MTT assay. Twenty-four healthy Wistar albino rats (150-200 g) of either sex were used for the in vivo investigations. Liver damage was induced by administration of 30% CCl4 suspended in olive oil (1 ml/kg body weight, i.p). RESULTS The antihepatotoxic effect of the total alkaloid fraction was observed in freshly isolated rat hepatocytes at very low concentrations (80-40 microg/ml). A dose-dependent increase in the percentage viability was observed when CCl4-exposed HepG2 cells were treated with different concentrations of the total alkaloid fraction. Its in vivo hepatoprotective effect at 80 mg/kg body weight was comparable with that of the standard Silymarin at 250 mg/kg body weight. CONCLUSION The total alkaloid fraction was able to normalize the biochemical levels which were altered due to CCl4 intoxication.
Collapse
Affiliation(s)
- Vasanth P Raj
- Manipal College of Pharmaceutical Sciences, Manipal University, Manipal - 576 104, India
| | | | | | | | | | | | | |
Collapse
|
38
|
Direct conversion of mouse fibroblasts to hepatocyte-like cells by defined factors. Nature 2011; 475:390-3. [PMID: 21716291 DOI: 10.1038/nature10263] [Citation(s) in RCA: 618] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2010] [Accepted: 06/06/2011] [Indexed: 02/06/2023]
Abstract
The location and timing of cellular differentiation must be stringently controlled for proper organ formation. Normally, hepatocytes differentiate from hepatic progenitor cells to form the liver during development. However, previous studies have shown that the hepatic program can also be activated in non-hepatic lineage cells after exposure to particular stimuli or fusion with hepatocytes. These unexpected findings suggest that factors critical to hepatocyte differentiation exist and become activated to induce hepatocyte-specific properties in different cell types. Here, by screening the effects of twelve candidate factors, we identify three specific combinations of two transcription factors, comprising Hnf4α plus Foxa1, Foxa2 or Foxa3, that can convert mouse embryonic and adult fibroblasts into cells that closely resemble hepatocytes in vitro. The induced hepatocyte-like (iHep) cells have multiple hepatocyte-specific features and reconstitute damaged hepatic tissues after transplantation. The generation of iHep cells may provide insights into the molecular nature of hepatocyte differentiation and potential therapies for liver diseases.
Collapse
|
39
|
Induction of a mature hepatocyte phenotype in adult liver derived progenitor cells by ectopic expression of transcription factors. Stem Cell Res 2011; 6:251-61. [PMID: 21474405 DOI: 10.1016/j.scr.2011.02.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Revised: 02/10/2011] [Accepted: 02/16/2011] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND/AIMS By ectopic expression of a distinct combination of transcription factors we aimed to induce a mature hepatocyte phenotype in an adult liver derived progenitor cell population (ALDPC). METHODS The open reading frames encoding murine Foxa2, Hnf4α and C/ebpα were cloned into lentivirus vectors and sequentially expressed in target cells. After seven days of culture, cells were analysed for expression of liver specific genes, and functional assays were performed. Fresh primary hepatocytes, twenty four hours in culture, served as positive controls. RESULTS Untransduced ALDPC under established differentiation conditions exhibited moderate signs of maturation, in particular in comparison with fresh hepatocyte controls. In transcription factor transduced cells, fifteen mRNA´s coding for secreted proteins, cytochrome p450 isoenzymes, liver metabolic enzymes were detected by RT-qPCR at levels close to controls. Albumin secretion increased incrementally in single (Foxa2), double (Foxa2, Hnf4α) and triple-transduced cells (Foxa2, Hnf4α, C/ebpα) and reached levels observed in primary hepatocytes. Glycogen storage as determined by PAS staining was detectable in double and triple transduced cells, comparable to controls. Ureagenesis was also induced in triple transduced cells, but at lower levels compared to primary hepatocytes. CONCLUSIONS Sequential expression of Foxa2, Hnf4α and C/ebpα induces a mature hepatocyte phenotype in an expandable liver derived progenitor cell line.
Collapse
|
40
|
Leconte I, Pallu S, Abarca-Quinones J, Michoux N, Peeters F, Radermacher K, Sempoux C, Najimi M, Sokal E, Van Beers BE. MRI of iron-oxide labelled transplanted hepatocytes in mice: effect of treatment with cyclophosphamide. J Magn Reson Imaging 2010; 32:367-75. [PMID: 20677264 DOI: 10.1002/jmri.22255] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
PURPOSE To assess if 1.5T MRI can be used to study the transport to the liver, the intrahepatic distribution and engraftment of iron-oxide labelled hepatocytes in cyclophosphamide-treated and untreated mice. MATERIALS AND METHODS Hepatocytes were isolated from C57bl/6 mice and were labelled with 1.63 microm iron-oxide particles. Seventeen mice were pretreated with cyclophosphamide to disrupt the sinusoidal endothelium and 15 were left untreated. Seven days after splenic injection of the labelled hepatocytes, T2*-weighted gradient-echo images at 1.5T were acquired. The hepatic transport, distribution and engraftment of the labelled hepatocytes were assessed with signal intensity (SI) and T2* measurements, electron paramagnetic resonance (EPR), texture analysis and histology. RESULTS Lower hepatic SI (P = 0.005), lower T2* (P = 0.033) and larger number of particles at histology (P = 0.006) suggested increased transport to the liver of labelled hepatocytes in cyclophosphamide-treated mice versus untreated mice. At histology, most particles were located in Kupffer cells. Particles distribution was heterogeneous. No difference between both groups was observed at texture analysis. CONCLUSION MRI is useful to assess the transport to the liver and intrahepatic distribution of transplanted labelled hepatocytes. The preferential location of iron-oxide particles within Kupffer cells after seven days limits the value of MRI for assessing hepatocyte engraftment.
Collapse
Affiliation(s)
- Isabelle Leconte
- Radiodiagnostic Unit, Université Catholique de Louvain, St-Luc University Hospital, Brussels, Belgium.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
|
42
|
Bitzegeio J, Bankwitz D, Hueging K, Haid S, Brohm C, Zeisel MB, Herrmann E, Iken M, Ott M, Baumert TF, Pietschmann T. Adaptation of hepatitis C virus to mouse CD81 permits infection of mouse cells in the absence of human entry factors. PLoS Pathog 2010; 6:e1000978. [PMID: 20617177 PMCID: PMC2895659 DOI: 10.1371/journal.ppat.1000978] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2010] [Accepted: 06/01/2010] [Indexed: 01/01/2023] Open
Abstract
Hepatitis C virus (HCV) naturally infects only humans and chimpanzees. The determinants responsible for this narrow species tropism are not well defined. Virus cell entry involves human scavenger receptor class B type I (SR-BI), CD81, claudin-1 and occludin. Among these, at least CD81 and occludin are utilized in a highly species-specific fashion, thus contributing to the narrow host range of HCV. We adapted HCV to mouse CD81 and identified three envelope glycoprotein mutations which together enhance infection of cells with mouse or other rodent receptors approximately 100-fold. These mutations enhanced interaction with human CD81 and increased exposure of the binding site for CD81 on the surface of virus particles. These changes were accompanied by augmented susceptibility of adapted HCV to neutralization by E2-specific antibodies indicative of major conformational changes of virus-resident E1/E2-complexes. Neutralization with CD81, SR-BI- and claudin-1-specific antibodies and knock down of occludin expression by siRNAs indicate that the adapted virus remains dependent on these host factors but apparently utilizes CD81, SR-BI and occludin with increased efficiency. Importantly, adapted E1/E2 complexes mediate HCV cell entry into mouse cells in the absence of human entry factors. These results further our knowledge of HCV receptor interactions and indicate that three glycoprotein mutations are sufficient to overcome the species-specific restriction of HCV cell entry into mouse cells. Moreover, these findings should contribute to the development of an immunocompetent small animal model fully permissive to HCV. The hepatitis C virus (HCV) infects only humans and chimpanzees, which has hampered development of suitable animal models. The inability of HCV to penetrate non-human cells is primarily due to inefficient usage of non-human CD81 and occludin. In this study we adapted HCV to mouse CD81. Efficient utilization of mouse CD81 is conferred by a combination of three mutations in the viral glycoproteins. These changes also permit entry via rat or hamster CD81, and lower viral dependence on additional HCV entry factors. Strikingly, mouse CD81 adapted HCV glycoproteins mediate entry into mouse cells in the absence of human entry factors. The adaptive mutations are not resident in viral domains implicated in direct CD81 binding. Nevertheless, they enhance binding to human CD81, increase susceptibility to different neutralizing antibodies and facilitate induction of viral cell fusion by low pH. This suggests that structural changes accompanied by exposure of the CD81 binding site and neutralizing epitopes have “unlocked” the viral envelope protein complex facilitating infection through non-human entry factors. These results highlight mechanisms of HCV receptor usage and tropism. They also demonstrate that HCV can be adapted to using non-human host factors, which may ultimately facilitate the development of small animal models.
Collapse
Affiliation(s)
- Julia Bitzegeio
- Division of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research; a joint venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Dorothea Bankwitz
- Division of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research; a joint venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Kathrin Hueging
- Division of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research; a joint venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Sibylle Haid
- Division of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research; a joint venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Christiane Brohm
- Division of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research; a joint venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | | | - Eva Herrmann
- Institute for Biostatistics and Mathematical Modeling, Johann Wolfgang Goethe-University, Frankfurt, Germany
| | - Marcus Iken
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Germany, and Clinical Research Group Cell and Gene Therapy, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Michael Ott
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Germany, and Clinical Research Group Cell and Gene Therapy, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | | | - Thomas Pietschmann
- Division of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research; a joint venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
- * E-mail:
| |
Collapse
|
43
|
Wahl M, Guenther R, Yang L, Bergman A, Straehle U, Strack S, Weiss C. Polybrominated diphenyl ethers and arylhydrocarbon receptor agonists: Different toxicity and target gene expression. Toxicol Lett 2010; 198:119-26. [PMID: 20566336 DOI: 10.1016/j.toxlet.2010.06.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2009] [Revised: 05/29/2010] [Accepted: 06/01/2010] [Indexed: 10/19/2022]
Abstract
Polybrominated diphenyl ethers (PBDEs) accumulate in the environment and in humans. PBDEs are developmental neurotoxicants, disturb the endocrine system and induce tumors in rodents. However, underlying mechanisms of PBDE toxicity are still insufficiently understood. Some reports demonstrated activation but also inhibition of the aryl hydrocarbon receptor (AhR) by PBDEs based on expression of its target gene cyp1A1. In the present study, we used different PBDE congeners (BDE47, 99, 153 and 209) and analyzed their effects on AhR signaling in various cell lines and zebrafish embryos. Furthermore, we performed microarray experiments in rat hepatoma cells to compare changes in gene expression induced by either BDE47 or the AhR agonist 2,3,7,8-tetrabromo-dibenzofuran (TBDF). PBDEs did not activate but rather inhibited AhR signaling and specifically induced malformations in zebrafish embryos, which differ from those provoked by AhR agonists. Furthermore, BDE47 and TBDF differentially regulated global gene expression in hepatoma cells. Hence, PBDEs and AhR agonists trigger different toxicity and target gene expression. Several novel target genes of BDE47 and TBDF were identified and verified by RT-PCR. TBDF induced expression of the transcriptional regulators Sim2 and RevErbbeta whereas BDE47 specifically deregulated expression of two subunits of the cytochrome c oxidase complex, cox6a2 and cox4i2, which might be linked to its toxicity.
Collapse
Affiliation(s)
- M Wahl
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology (KIT), Campus North, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | | | | | | | | | | | | |
Collapse
|
44
|
Mazzei D, Guzzardi MA, Giusti S, Ahluwalia A. A low shear stress modular bioreactor for connected cell culture under high flow rates. Biotechnol Bioeng 2010; 106:127-37. [PMID: 20091740 DOI: 10.1002/bit.22671] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
A generic "system on a plate" modular multicompartmental bioreactor array which enables microwell protocols to be transferred directly to the bioreactor modules, without redesign of cell culture experiments or protocols is described. The modular bioreactors are simple to assemble and use and can be easily compared with standard controls since cell numbers and medium volumes are quite similar. Starting from fluid dynamic and mass transport considerations, a modular bioreactor chamber was first modeled and then fabricated using "milli-molding," a technique adapted from soft lithography. After confirming that the shear stress was extremely low in the system in the range of useful flow rates, the bioreactor chambers were tested using hepatocytes. The results show that the bioreactor chambers can increase or maintain cell viability and function when the flow rates are below 500 microL/min, corresponding to wall shear stresses of 10(-5) Pa or less at the cell culture surface.
Collapse
Affiliation(s)
- D Mazzei
- Faculty of Engineering, Interdepartmental Research Center E. Piaggio, University of Pisa, Pisa, Italy.
| | | | | | | |
Collapse
|
45
|
Sakuma S, Nakanishi M, Morinaga K, Fujitake M, Wada SI, Fujimoto Y. Bisphenol A 3,4-quinone induces the conversion of xanthine dehydrogenase into oxidase in vitro. Food Chem Toxicol 2010; 48:2217-22. [PMID: 20594952 DOI: 10.1016/j.fct.2010.05.051] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2009] [Revised: 04/28/2010] [Accepted: 05/18/2010] [Indexed: 12/23/2022]
Abstract
In the present study, we assessed the influence of bisphenol A (BPA) and bisphenol A 3,4-quinone (BPAQ) on the conversion of xanthine dehydrogenase (XD) into xanthine oxidase (XO) in the rat liver in vitro. BPA up to 100 micromol/L did not affect the XO and XD activities in the partially purified cytosolic fraction from rat liver, whereas BPAQ (2-10 micromol/L) dose-dependently enhanced the XO activity concomitant with a decrease in the XD activity, implying that BPAQ, but not BPA, can convert XD into the reactive oxygen species (ROS) producing the form XO. Furthermore, it was found that BPAQ could increase the generation of ROS and oxidize the guanine moiety of deoxyguanosine in the DNA of primary rat hepatocyte cultures. These results suggest that BPAQ has the potential to convert XD into XO in the liver, which in turn may lead to ROS generation and oxidative DNA damage in this region.
Collapse
Affiliation(s)
- Satoru Sakuma
- Laboratory of Physiological Chemistry, Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka 569-1094, Japan.
| | | | | | | | | | | |
Collapse
|
46
|
Uygun BE, Bou-Akl T, Albanna M, Matthew HWT. Membrane thickness is an important variable in membrane scaffolds: Influence of chitosan membrane structure on the behavior of cells. Acta Biomater 2010; 6:2126-31. [PMID: 19925888 DOI: 10.1016/j.actbio.2009.11.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2009] [Revised: 10/13/2009] [Accepted: 11/11/2009] [Indexed: 01/08/2023]
Abstract
Cell and tissue responses to polymeric materials are orchestrated in part by the conformations of adsorbed plasma proteins. Thus, the chemical properties of a polymer membrane that govern protein adsorption behavior can play an important role in determining the biological properties of tissue engineered scaffolds derived from that polymer. In this study, we explored the role of membrane thickness as a factor influencing cell adhesion and proliferation on chitosan membranes with and without covalently attached glycosaminoglycans. Rat mesenchymal stem cells (MSCs) cultured on chitosan membranes of various thicknesses demonstrated significantly improved cell adhesion, spreading and proliferation as membrane thickness was increased. Rat hepatocytes displayed increased spreading on the substrate with increasing membrane thickness, similar to MSCs. Increased thickness reduced the overall crystallinity of the membrane, and the data indicate that the improved cellular responses were likely due to enhanced adsorption of serum vitronectin, presumably due to reduced membrane crystallinity. These results demonstrate that membrane thickness is an important design variable that can be manipulated in chitosan-based scaffolds to achieve enhanced cell spreading, proliferation and function.
Collapse
Affiliation(s)
- Basak E Uygun
- Department of Chemical Engineering and Materials Science, Wayne State University, 5050 Anthony Wayne Drive, Detroit, MI 48202, USA
| | | | | | | |
Collapse
|
47
|
Wallace K, Fairhall EA, Charlton KA, Wright MC. AR42J-B-13 cell: an expandable progenitor to generate an unlimited supply of functional hepatocytes. Toxicology 2010; 278:277-87. [PMID: 20685382 DOI: 10.1016/j.tox.2010.05.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2010] [Revised: 05/17/2010] [Accepted: 05/18/2010] [Indexed: 01/16/2023]
Abstract
Hepatocytes are the preparation of choice for Toxicological research in vitro. However, despite the fact that hepatocytes proliferate in vivo during liver regeneration, they are resistant to proliferation in vitro, do not tolerate sub-culture and tend to enter a de-differentiation program that results in a loss of hepatic function. These limitations have resulted in the search for expandable rodent and human cells capable of being directed to differentiate into functional hepatocytes. Research with stem cells suggests that it may be possible to provide the research community with hepatocytes in vitro although to date, significant challenges remain, notably generating a sufficiently pure population of hepatocytes with a quantitative functionality comparable with hepatocytes. This paper reviews work with the AR42J-B-13 (B-13) cell line. The B-13 cell was cloned from the rodent AR42J pancreatic cell line, express genes associated with pancreatic acinar cells and readily proliferates in simple culture media. When exposed to glucocorticoid, 75-85% of the cells trans-differentiate into hepatocyte-like (B-13/H) cells functioning at a level quantitatively similar to freshly isolated rat hepatocytes (with the remaining cells retaining the B-13 phenotype). Trans-differentiation of pancreatic acinar cells also appears to occur in vivo in rats treated with glucocorticoid; in mice with elevated circulating glucocorticoid and in humans treated for long periods with glucocorticoid. The B-13 response to glucocorticoid therefore appears to be related to a real pathophysiological response of a pancreatic cell to glucocorticoid. An understanding of how this process occurs and if it can be generated or engineered in human cells would result in a cell line with the ability to generate an unlimited supply of functional human hepatocytes in a cost effective manner.
Collapse
Affiliation(s)
- Karen Wallace
- Institute of Cellular Medicine, Medical School, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | | | | | | |
Collapse
|
48
|
Lohwasser C, Neureiter D, Popov Y, Bauer M, Schuppan D. Role of the receptor for advanced glycation end products in hepatic fibrosis. World J Gastroenterol 2009; 15:5789-98. [PMID: 19998499 PMCID: PMC2791271 DOI: 10.3748/wjg.15.5789] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To study the role of advanced glycation end products (AGE) and their specific receptor (RAGE) in the pathogenesis of liver fibrogenesis.
METHODS: In vitro RAGE expression and extracellular matrix-related gene expression in both rat and human hepatic stellate cells (HSC) were measured after stimulation with the two RAGE ligands, advanced glycation end product-bovine serum albumin (AGE-BSA) and Nε-(carboxymethyl) lysine (CML)-BSA, or with tumor necrosis factor-α (TNF-α). In vivo RAGE expression was examined in models of hepatic fibrosis induced by bile duct ligation or thioacetamide. The effects of AGE-BSA and CML-BSA on HSC proliferation, signal transduction and profibrogenic gene expression were studied in vitro.
RESULTS: In hepatic fibrosis, RAGE expression was enhanced in activated HSC, and also in endothelial cells, inflammatory cells and activated bile duct epithelia. HSC expressed RAGE which was upregulated after stimulation with AGE-BSA, CML-BSA, and TNF-α. RAGE stimulation with AGE-BSA and CML-BSA did not alter HSC proliferation, apoptosis, fibrogenic signal transduction and fibrosis- or fibrolysis-related gene expression, except for marginal upregulation of procollagen α1(I) mRNA by AGE-BSA.
CONCLUSION: Despite upregulation of RAGE in activated HSC, RAGE stimulation by AGE does not alter their fibrogenic activation. Therefore, RAGE does not contribute directly to hepatic fibrogenesis.
Collapse
|
49
|
Poirier A, Cascais AC, Funk C, Lavé T. Prediction of pharmacokinetic profile of valsartan in human based on in vitro uptake transport data. J Pharmacokinet Pharmacodyn 2009; 36:585-611. [DOI: 10.1007/s10928-009-9139-3] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2009] [Accepted: 11/07/2009] [Indexed: 12/21/2022]
|
50
|
Deurholt T, van Til NP, Chhatta AA, ten Bloemendaal L, Schwartlander R, Payne C, Plevris JN, Sauer IM, Chamuleau RA, Elferink RPO, Seppen J, Hoekstra R. Novel immortalized human fetal liver cell line, cBAL111, has the potential to differentiate into functional hepatocytes. BMC Biotechnol 2009; 9:89. [PMID: 19845959 PMCID: PMC2770505 DOI: 10.1186/1472-6750-9-89] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2008] [Accepted: 10/21/2009] [Indexed: 01/15/2023] Open
Abstract
Background A clonal cell line that combines both stable hepatic function and proliferation capacity is desirable for in vitro applications that depend on hepatic function, such as pharmacological or toxicological assays and bioartificial liver systems. Here we describe the generation and characterization of a clonal human cell line for in vitro hepatocyte applications. Results Cell clones derived from human fetal liver cells were immortalized by over-expression of telomerase reverse transcriptase. The resulting cell line, cBAL111, displayed hepatic functionality similar to the parental cells prior to immortalization, and did not grow in soft agar. Cell line cBAL111 expressed markers of immature hepatocytes, like glutathione S transferase and cytokeratin 19, as well as progenitor cell marker CD146 and was negative for lidocaine elimination. On the other hand, the cBAL111 cells produced urea, albumin and cytokeratin 18 and eliminated galactose. In contrast to hepatic cell lines NKNT-3 and HepG2, all hepatic functions were expressed in cBAL111, although there was considerable variation in their levels compared with primary mature hepatocytes. When transplanted in the spleen of immunodeficient mice, cBAL111 engrafted into the liver and partly differentiated into hepatocytes showing expression of human albumin and carbamoylphosphate synthetase without signs of cell fusion. Conclusion This novel liver cell line has the potential to differentiate into mature hepatocytes to be used for in vitro hepatocyte applications.
Collapse
Affiliation(s)
- Tanja Deurholt
- AMC Liver Center, Meibergdreef 69-71, 1105 BK Amsterdam, the Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|