1
|
Funahashi N, Okada H, Kaneko R, Nio K, Yamashita T, Koshikawa N. Hepatocyte transformation is induced by laminin γ2 monomer. Cancer Sci 2024; 115:2972-2984. [PMID: 38951133 PMCID: PMC11462950 DOI: 10.1111/cas.16265] [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: 02/26/2024] [Revised: 06/15/2024] [Accepted: 06/20/2024] [Indexed: 07/03/2024] Open
Abstract
Serum laminin-γ2 monomer (Lm-γ2m) is a potent predictive biomarker for hepatocellular carcinoma (HCC) onset in patients with hepatitis C infection who achieve a sustained virologic response with liver cirrhosis (LC) and for the onset of extrahepatic metastases in early-stage HCC. Although Lm-γ2m involvement in late-stage cancer progression has been well investigated, its precise roles in HCC onset remain to be systematically investigated. Therefore, we analyzed an HCC model, human hepatocytes and cholangiocytes, and surgically resected liver tissues from patients with HCC to understand the roles of Lm-γ2m in HCC onset. Ck-19- and EpCAM-positive hepatic progenitor cells (HPCs) in the liver of pdgf-c transgenic HCC mouse model with ductular reaction showed ectopic expression of Lm-γ2m. Forced expression of Lm-γ2m in hepatocytes adjacent to HPCs resulted in enhanced tumorigenicity, cell proliferation, and migration in immortalized hepatocytes, but not in cholangiocytes in vitro. Further, pharmacological inhibition of epidermal growth factor receptor (EGFR) and c-Jun activator JNK suppressed Lm-γ2m-induced hepatocyte transformation, suggesting the involvement of EGFR/c-Jun signaling in the transformation, leading to HCC development. Finally, immunohistochemical staining of HCC tissues revealed a high level of Lm-γ2 expression in the HPCs of the liver with ductular reaction in normal liver adjacent to HCC tissues. Overall, HPC-derived Lm-γ2m in normal liver with ductular reaction acts as a paracrine growth factor on surrounding hepatocytes and promotes their cellular transformation through the EGFR/c-Jun signaling pathway. Furthermore, this is the first report on Lm-γ2m expression detected in the normal liver with ductular reaction, a human precancerous lesion of HCC.
Collapse
Affiliation(s)
- Nobuaki Funahashi
- Department of Life Science and TechnologyTokyo Institute of TechnologyYokohamaKanagawaJapan
| | - Hikari Okada
- Department of Gastroenterology, Graduate School of Medical ScienceKanazawa UniversityKanazawaIshikawaJapan
| | - Ryo Kaneko
- Department of Life Science and TechnologyTokyo Institute of TechnologyYokohamaKanagawaJapan
| | - Kouki Nio
- Department of Gastroenterology, Graduate School of Medical ScienceKanazawa UniversityKanazawaIshikawaJapan
| | - Taro Yamashita
- Department of Gastroenterology, Graduate School of Medical ScienceKanazawa UniversityKanazawaIshikawaJapan
| | - Naohiko Koshikawa
- Department of Life Science and TechnologyTokyo Institute of TechnologyYokohamaKanagawaJapan
- Clinical Cancer Proteomics LaboratoryKanagawa Cancer Center Research InstituteYokohamaKanagawaJapan
| |
Collapse
|
2
|
Bi YH, Han WQ, Li RF, Wang YJ, Du ZS, Wang XJ, Jiang Y. Signal transducer and activator of transcription 3 promotes the Warburg effect possibly by inducing pyruvate kinase M2 phosphorylation in liver precancerous lesions. World J Gastroenterol 2019; 25:1936-1949. [PMID: 31086462 PMCID: PMC6487376 DOI: 10.3748/wjg.v25.i16.1936] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 03/07/2019] [Accepted: 03/25/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Study shows that signal transducer and activator of transcription 3 (STAT3) can increase the Warburg effect by stimulating hexokinase 2 in breast cancer and upregulate lactate dehydrogenase A and pyruvate dehydrogenase kinase 1 in myeloma. STAT3 and pyruvate kinase M2 (PKM2) can also be activated and enhance the Warburg effect in hepatocellular carcinoma. Precancerous lesions are critical to human and rodent hepatocarcinogenesis. However, the underlying molecular mechanism for the development of liver precancerous lesions remains unknown. We hypothesized that STAT3 promotes the Warburg effect possibly by upregulating p-PKM2 in liver precancerous lesions in rats.
AIM To investigate the mechanism of the Warburg effect in liver precancerous lesions in rats.
METHODS A model of liver precancerous lesions was established by a modified Solt-Farber method. The liver pathological changes were observed by HE staining and immunohistochemistry. The transformation of WB-F344 cells induced with N-methyl-N’-nitro-N-nitrosoguanidine and hydrogen peroxide was evaluated by the soft agar assay and aneuploidy. The levels of glucose and lactate in the tissue and culture medium were detected with a spectrophotometer. The protein levels of glutathione S-transferase-π, proliferating cell nuclear antigen (PCNA), STAT3, and PKM2 were examined by Western blot and immunofluorescence.
RESULTS We found that the Warburg effect was increased in liver precancerous lesions in rats. PKM2 and p-STAT3 were upregulated in activated oval cells in liver precancerous lesions in rats. The Warburg effect, p-PKM2, and p-STAT3 expression were also increased in transformed WB-F344 cells. STAT3 activation promoted the clonal formation rate, aneuploidy, alpha-fetoprotein expression, PCNA expression, G1/S phase transition, the Warburg effect, PKM2 phosphorylation, and nuclear translocation in transformed WB-F344 cells. Moreover, the Warburg effect was inhibited by stattic, a specific inhibitor of STAT3, and further reduced in transformed WB-F344 cells after the intervention for PKM2.
CONCLUSION The Warburg effect is initiated in liver precancerous lesions in rats. STAT3 activation promotes the Warburg effect by enhancing the phosphorylation of PKM2 in transformed WB-F344 cells.
Collapse
Affiliation(s)
- Yang-Hui Bi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Wen-Qi Han
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Ruo-Fei Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Yun-Jiao Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Zun-Shu Du
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Xue-Jiang Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Ying Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| |
Collapse
|
3
|
TGF-β1 Induces the Dual Regulation of Hepatic Progenitor Cells with Both Anti- and Proliver Fibrosis. Stem Cells Int 2015; 2016:1492694. [PMID: 26839553 PMCID: PMC4709730 DOI: 10.1155/2016/1492694] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 09/17/2015] [Indexed: 02/06/2023] Open
Abstract
Transforming growth factor-beta 1 (TGF-β1) plays a central role in hepatic progenitor cells- (HPCs-) mediated liver repair and fibrosis. However, different effects of TGF-β1 on progenitor cells have not been described. In this study, both in vitro (HPCs cocultured with hepatic stellate cells (HSCs) in transwells) and in vivo (CCl4-injured liver fibrosis rat) systems were used to evaluate the impacts. We found that HPCs pretreated with TGF-β1 for 12 hours inhibited the activation of HSCs, while sensitization for 48 hours increased the activation of HSCs. Consistent with these in vitro results, the in vivo fibrosis rat model showed the same time-dependent dual effect of TGF-β1. Regression of liver fibrosis as well as normalization of serum aminotransferase and albumin levels was detected in the rats transplanted with HPCs pretreated with TGF-β1 for 12 hours. In contrast, severe liver fibrosis and elevated collagen-1 levels were detected in the rats transplanted with HPCs pretreated with TGF-β1 for 48 hours. Furthermore, the TGF-β1-pretreated HPCs were shown to deactivate HSCs via enhancing SERPINE1 expression. Inhibition of SERPINE1 reversed the deactivation response in a dose-dependent manner.
Collapse
|
4
|
Zhang P, Zhu X, Wu Y, Hu R, Li D, Du J, Jiao X, He X. Histone deacetylase inhibitors reduce WB-F344 oval cell viability and migration capability by suppressing AKT/mTOR signaling in vitro. Arch Biochem Biophys 2015; 590:1-9. [PMID: 26558695 DOI: 10.1016/j.abb.2015.11.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 11/03/2015] [Accepted: 11/04/2015] [Indexed: 12/12/2022]
Abstract
Histone deacetylase (HDAC) can blockDNA replication and transcription and altered HDAC expression was associated with tumorigenesis. This study investigated the effects of HDAC inhibitors on hepatic oval cells and aimed to delineate the underlying molecular events. Hepatic oval cells were treated with two different HDAC inhibitors, suberoylanilidehydroxamic acid (SAHA) and trichostatin-A (TSA). Cells were subjected to cell morphology, cell viability, cell cycle, and wound healing assays. The expression of proteins related to both apoptosis and the cell cycle, and proteins of the AKT/mammalian target of rapamycin (mTOR) signaling pathway were analyzed by Western blot. The data showed that HDAC inhibitors reduced oval cell viability and migration capability, and arrested oval cells at the G0/G1 and S phases of the cell cycle, in a dose- and time-dependent manner. HDAC inhibitors altered cell morphology and reduced oval cell viability, and downregulated the expression of PCNA, cyclinD1, c-Myc and Bmi1 proteins, while also suppressing AKT/mTOR and its downstream target activity. In conclusion, this study demonstrates that HDAC inhibitors affect oval cells by suppressing AKT/mTOR signaling.
Collapse
Affiliation(s)
- Peng Zhang
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiaofeng Zhu
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ying Wu
- Department of Biostatistics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ronglin Hu
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Dongming Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jun Du
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xingyuan Jiao
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Xiaoshun He
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
5
|
He Y, Cui J, He T, Bi Y. 5-azacytidine promotes terminal differentiation of hepatic progenitor cells. Mol Med Rep 2015; 12:2872-8. [PMID: 25975647 DOI: 10.3892/mmr.2015.3772] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 03/24/2015] [Indexed: 11/06/2022] Open
Abstract
5-azacytidine (5-azaC) is known to induce cardiomyocyte differentiation. However, its function in hepatocyte differentiation is unclear. The present study investigated the in vitro capability of 5-azaC to promote maturation and differentiation of mouse embryonic hepatic progenitor cells, with the aim of developing an approach for improving hepatic differentiation. Mouse embryonic hepatic progenitor cells (HP14.5 cells) were treated with 5-azaC at concentrations from 0 to 20 μmol/l, in addition to hepatocyte induction culture medium. Hepatocyte induction medium induces HP14.5 cell differentiation. 5-azaC may enhance the albumin promotor-driven Gaussia luciferase (ALB-GLuc) activity in induced HP14.5 cells. In the present study 2 μmol/l was found to be the optimum concentration with which to achieve this. The expression of hepatocyte-associated factors was not significantly different between the group treated with 5-azaC alone and the control group. The mRNA levels of ALB; cytokeratin 18 (CK18); tyrosine aminotransferase (TAT); and cytochrome p450, family 1, member A1 (CYP1A1); in addition to the protein levels of ALB, CK18 and uridine diphosphate glucuronyltransferase 1A (UGT1A) in the induced group with 5-azaC, were higher than those in the induced group without 5-azaC, although no significant differences were detected in expression of the hepatic stem cell markers, DLK and α-fetoprotein, between the two groups. Treatment with 5-azaC alone did not affect glycogen synthesis or indocyanine green (ICG) metabolic function in HP14.5 cells, although it significantly increased ICG uptake and periodic acid-Schiff-positive cell numbers amongst HP14.5 cells. Therefore, the present study demonstrated that treatment with 5-azaC alone exerted no effects on the maturation and differentiation of HP14.5 cells. However, 5-azaC exhibited a synergistic effect on the terminal differentiation of induced hepatic progenitor cells in association with a hepatic induction medium.
Collapse
Affiliation(s)
- Yun He
- Department of Pediatric Surgery, The Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
| | - Jiejie Cui
- Stem Cell Biology and Therapy Laboratory, The Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
| | - Tongchuan He
- Stem Cell Biology and Therapy Laboratory, The Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
| | - Yang Bi
- Department of Pediatric Surgery, The Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
| |
Collapse
|
6
|
Khademi F, Soleimani M, Verdi J, Tavangar SM, Sadroddiny E, Masumi M, Ai J. Human endometrial stem cells differentiation into functional hepatocyte-like cells. Cell Biol Int 2014; 38:825-34. [DOI: 10.1002/cbin.10278] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Accepted: 02/07/2014] [Indexed: 12/30/2022]
Affiliation(s)
- Farzaneh Khademi
- Department of Tissue Engineering; School of Advanced Technologies in Medicine; Tehran University of Medical Sciences; Tehran Iran
- Stem Cells Technology Research Center; Tehran Iran
| | - Masoud Soleimani
- Stem Cells Technology Research Center; Tehran Iran
- Department of Hematology; Faculty of Medical Science; Tarbiat Modares University; Tehran Iran
| | - Javad Verdi
- Department of Tissue Engineering; School of Advanced Technologies in Medicine; Tehran University of Medical Sciences; Tehran Iran
- Department of Applied Cell; School of Advanced Technologies in Medicine; Tehran University of Medical Sciences; Tehran Iran
| | - Seyed Mohammad Tavangar
- Department of Tissue Engineering; School of Advanced Technologies in Medicine; Tehran University of Medical Sciences; Tehran Iran
- Department of Pathology; Shariaty Hospital; Tehran University of Medical Sciences; Tehran Iran
| | - Esmaeil Sadroddiny
- Department of Medical Biotechnology; School of Advanced Technologies in Medicine; Tehran University of Medical Sciences; Tehran Iran
| | - Mohammad Masumi
- Stem Cells Technology Research Center; Tehran Iran
- Induced Pluripotent Stem Cell Biotechnology Team; Stem Cells Department; National Institute of Genetic Engineering and Biotechnology; Tehran Iran
| | - Jafar Ai
- Department of Tissue Engineering; School of Advanced Technologies in Medicine; Tehran University of Medical Sciences; Tehran Iran
- Brain and Spinal Injury Research Center; Tehran University of Medical Sciences; Tehran Iran
| |
Collapse
|
7
|
Chen L, Zhang W, Liang HF, Zhou QD, Ding ZY, Yang HQ, Liu WB, Wu YH, Man Q, Zhang BX, Chen XP. Activin A induces growth arrest through a SMAD- dependent pathway in hepatic progenitor cells. Cell Commun Signal 2014; 12:18. [PMID: 24628936 PMCID: PMC3995548 DOI: 10.1186/1478-811x-12-18] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 03/08/2014] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Activin A, an important member of transforming growth factor-β superfamily, is reported to inhibit proliferation of mature hepatocyte. However, the effect of activin A on growth of hepatic progenitor cells is not fully understood. To that end, we attempted to evaluate the potential role of activin A in the regulation of hepatic progenitor cell proliferation. RESULTS Using the 2-acetaminofluorene/partial hepatectomy model, activin A expression decreased immediately after partial hepatectomy and then increased from the 9th to 15th day post surgery, which is associated with the attenuation of oval cell proliferation. Activin A inhibited oval cell line LE6 growth via activating the SMAD signaling pathway, which manifested as the phosphorylation of SMAD2/3, the inhibition of Rb phosphorylation, the suppression of cyclinD1 and cyclinE, and the promotion of p21WAF1/Cip1 and p15INK4B expression. Treatment with activin A antagonist follistatin or blocking SMAD signaling could diminish the anti-proliferative effect of activin A. By contrast, inhibition of the MAPK pathway did not contribute to this effect. Antagonizing activin A activity by follistatin administration enhanced oval cell proliferation in the 2-acetylaminofluorene/partial hepatectomy model. CONCLUSION Activin A, acting through the SMAD pathway, negatively regulates the proliferation of hepatic progenitor cells.
Collapse
Affiliation(s)
- Lin Chen
- Hepatic surgery centre, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Zhang
- Hepatic surgery centre, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Hui-fang Liang
- Hepatic surgery centre, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Qiao-dan Zhou
- Department of Nephrology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Ze-yang Ding
- Hepatic surgery centre, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Hong-qiang Yang
- Hepatic surgery centre, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Medical College of Shihezi University, Shihezi, China
| | - Wei-bo Liu
- Hepatic surgery centre, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yan-hui Wu
- Hepatic surgery centre, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Quan Man
- Hepatic surgery centre, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Bi-xiang Zhang
- Hepatic surgery centre, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-ping Chen
- Hepatic surgery centre, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
8
|
Bi Y, He Y, Huang JY, Xu L, Tang N, He TC, Feng T. Induced maturation of hepatic progenitor cells in vitro. Braz J Med Biol Res 2013; 46:559-66. [PMID: 23903683 PMCID: PMC3859339 DOI: 10.1590/1414-431x20132455] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2012] [Accepted: 04/15/2013] [Indexed: 12/18/2022] Open
Abstract
Hepatic progenitor cells (HPCs) are a potential cell source for liver cell
transplantation but do not function like mature liver cells. We sought an
effective and reliable method to induce HPC maturation. An immortalized HP14.5
albumin promoter-driven Gaussian luciferase (ALB-GLuc) cell line was established
from HPCs isolated from fetal mouse liver of post coitus day 14.5 mice to
investigate the effect of induction factors on ALB promoter. HP14.5 parental
cells were cultured in DMEM with different combinations of 2% horse serum (HS),
0.1 µM dexamethasone (DEX), 10 ng/mL hepatic growth factor (HGF), and/or 20
ng/mL fibroblast growth factor 4 (FGF4). Trypan blue and crystal violet staining
were used to assess cell proliferation with different induction conditions.
Expression of hepatic markers was measured by semi-quantitative RT-PCR, Western
blot, and immunofluorescence. Glycogen storage and metabolism were detected by
periodic acid-Schiff and indocyanine green (ICG) staining. GLuc activity
indicated ALB expression. The combination of 2% HS+0.1 µM Dex+10 ng/mL HGF+20
ng/mL FGF4 induced the highest ALB-GLuc activity. Cell proliferation decreased
in 2% HS but increased by adding FGF4. Upon induction, and consistent with
hepatocyte development, DLK, AFP, and CK19 expression decreased, while ALB,
CK18, and UGT1A expression increased. The maturity markers tyrosine
aminotransferase and apolipoprotein B were detected at days 3 and 6
post-induction, respectively. ICG uptake and glycogen synthesis were detectable
at day 6 and increased over time. Therefore, we demonstrated that HPCs were
induced to differentiate into functional mature hepatocytes in
vitro, suggesting that factor-treated HPCs may be further explored
as a means of liver cell transplantation.
Collapse
Affiliation(s)
- Y Bi
- Stem Cell Biology and Therapy Laboratory, Department of Pediatric Surgery, Key Laboratory of Child Development and Disorders, Ministry of Education, Chongqing, China
| | | | | | | | | | | | | |
Collapse
|
9
|
Fetal liver cell transplantation as a potential alternative to whole liver transplantation? J Gastroenterol 2011; 46:953-65. [PMID: 21698354 DOI: 10.1007/s00535-011-0427-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Accepted: 05/02/2011] [Indexed: 02/04/2023]
Abstract
Because organ shortage is the fundamental limitation of whole liver transplantation, novel therapeutic options, especially the possibility of restoring liver function through cell transplantation, are urgently needed to treat end-stage liver diseases. Groundbreaking in vivo studies have shown that transplanted hepatocytes are capable of repopulating the rodent liver. The two best studied models are the urokinase plasminogen activator (uPA) transgenic mouse and the fumarylacetoacetate hydrolase (FAH)-deficient mouse, in which genetic modifications of the recipient liver provide a tissue environment in which there is extensive liver injury and selection pressure favoring the proliferation and survival of transplanted hepatocytes. Because transplanted hepatocytes do not significantly repopulate the (near-)normal liver, attention has been focused on finding alternative cell types, such as stem or progenitor cells, that have a higher proliferative potential than hepatocytes. Several sources of stem cells or stem-like cells have been identified and their potential to repopulate the recipient liver has been evaluated in certain liver injury models. However, rat fetal liver stem/progenitor cells (FLSPCs) are the only cells identified to date that can effectively repopulate the (near-)normal liver, are morphologically and functionally fully integrated into the recipient liver, and remain viable long-term. Even though primary human fetal liver cells are not likely to be routinely used for clinical liver cell repopulation in the future, using or engineering candidate cells exhibiting the characteristics of FLSPCs suggests a new direction in developing cell transplantation strategies for therapeutic liver replacement. This review will give a brief overview concerning the existing animal models and cell sources that have been used to restore normal liver structure and function, and will focus specifically on the potential of FLSPCs to repopulate the liver.
Collapse
|
10
|
Abstract
In 1938, the field of Transfusion Medicine began as the simpler entity - Blood Banking. It was a discipline that focused on collecting, processing, storing and distributing end stage blood cells, plasma and plasma fractions to patients. Over the years, the field progressed to include clinical patient services such as apheresis technology and with the development of stem cell transplantation as a standard of care, Cell Therapy. Now the discipline is also finding a niche in the area of Regenerative Medicine. The role played by Transfusion Medicine practitioners in Cell Therapy and Regenerative Medicine was predicated on many factors: (1) pre-existing, established protocols for therapeutic leukapheresis, (2) prior experience with mononuclear cell collection and processing, (3) long term familiarity with, and a clear understanding of, cGMP and cGLP guidelines, Federal regulations, and the voluntary standards established by various organizations, (4) close relationships with practitioners in clinical departments of medicine, pediatrics, oncology, surgery, and their subspecialty areas. While the initial Cell Therapy programs related primarily to hematopoietic stem cell transplantation, as Regenerative Medicine programs developed, transfusion specialists found it to be a related field that would also benefit from their input. Cell Therapy and Regenerative Medicine, now provide fertile soil for the seeds of Transfusion Medicine to grow. The once narrowly defined field of Blood Banking now encompasses involvement in major new Cellular Therapy/Regenerative Medicine research protocols related to treatment of patients with cancer as well as renal, hepatic and cardiac illnesses. This in turn provides opportunities for residents and fellows to consider robust careers in the field of Transfusion Medicine. In this manner we will move forward with one eye on the past and another on the promising future.
Collapse
Affiliation(s)
- Jacquelyn Choate
- Department of Laboratory Medicine, Yale University School of Medicine, CT, USA
| | | |
Collapse
|
11
|
Szidonya J, Farkas T, Pali T. The fatty acid constitution and ordering state of membranes in dominant temperature-sensitive lethal mutation and wild-type Drosophila melanogaster larvae. Biochem Genet 1990; 5:26-32. [PMID: 2168167 DOI: 10.1007/s11684-011-0107-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2010] [Accepted: 12/23/2010] [Indexed: 01/11/2023]
Abstract
The ordering state and changes in fatty acid composition of microsomal (MS) and mitochondrial (MC) membranes of two dominant temperature-sensitive (DTS) lethal mutations and the wild-type Oregon-R strain larvae of Drosophila melanogaster have been studied at 18 and 29 degrees C and after temperature-shift experiments. The membranes of wild-type larvae have a stable ordering state, with "S" values between 0.6 (18 degrees C) and 0.5 (29 degrees C) in both membranes which remained unchanged in shift experiments, although the ratios of saturated/unsaturated fatty acids were changed as expected. The strongly DTS mutation 1(2) 10DTS forms very rigid membranes at the restrictive temperature (29 degrees C) which cannot be normalized after shift down, while shift up or development at the permissive temperature results in normal ordering state. This mutant is less able to adjust MS and MC fatty acid composition in response to the growth temperature than the wild type. The less temperature-sensitive 1(2)2DTS allele occupies an intermediate state between Oregon-R and 1(2)10DTS in both respects. We assume and the genetical data suggest that the DTS mutant gene product is in competition with the wild-type product, resulting in a membrane structure which is not able to accommodate to the restrictive temperature.
Collapse
Affiliation(s)
- J Szidonya
- Institute of Genetics, Hungarian Academy of Sciences, Szeged
| | | | | |
Collapse
|