1
|
Corton JC, Ledbetter V, Cohen SM, Atlas E, Yauk CL, Liu J. A transcriptomic biomarker predictive of cell proliferation for use in adverse outcome pathway-informed testing and assessment. Toxicol Sci 2024; 201:174-189. [PMID: 39137154 DOI: 10.1093/toxsci/kfae102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024] Open
Abstract
High-throughput transcriptomics (HTTr) is increasingly being used to identify molecular targets of chemicals that can be linked to adverse outcomes. Cell proliferation (CP) is an important key event in chemical carcinogenesis. Here, we describe the construction and characterization of a gene expression biomarker that is predictive of the CP status in human and rodent tissues. The biomarker was constructed from 30 genes known to be increased in expression in prostate cancers relative to surrounding tissues and in cycling human MCF-7 cells after estrogen receptor (ER) agonist exposure. Using a large compendium of gene expression profiles to test utility, the biomarker could identify increases in CP in (i) 308 out of 367 tumor vs. normal surrounding tissue comparisons from 6 human organs, (ii) MCF-7 cells after activation of ER, (iii) after partial hepatectomy in mice and rats, and (iv) the livers of mice and rats after exposure to nongenotoxic hepatocarcinogens. The biomarker identified suppression of CP (i) under conditions of p53 activation by DNA damaging agents in human cells, (ii) in human A549 lung cells exposed to therapeutic anticancer kinase inhibitors (dasatinib, nilotnib), and (iii) in the mouse liver when comparing high levels of CP at birth to the low background levels in the adult. The responses using the biomarker were similar to those observed using conventional markers of CP including PCNA, Ki67, and BrdU labeling. The CP biomarker will be a useful tool for interpretation of HTTr data streams to identify CP status after exposure to chemicals in human cells or in rodent tissues.
Collapse
Affiliation(s)
- J Christopher Corton
- Center for Computational Toxicology and Exposure, US Environmental Protection Agency, Research Triangle Park, NC 27711, United States
| | - Victoria Ledbetter
- Center for Computational Toxicology and Exposure, US Environmental Protection Agency, Research Triangle Park, NC 27711, United States
| | - Samuel M Cohen
- Department of Pathology and Microbiology and Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 69198-3135, United States
| | - Ella Atlas
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch (HECSB) Health Canada, Ottawa, ON K2K 0K9, Canada
| | - Carole L Yauk
- Department of Biology, University of Ottawa, Ottawa, ON, Canada
| | - Jie Liu
- Center for Computational Toxicology and Exposure, US Environmental Protection Agency, Research Triangle Park, NC 27711, United States
| |
Collapse
|
2
|
Nishikawa Y. Aberrant differentiation and proliferation of hepatocytes in chronic liver injury and liver tumors. Pathol Int 2024; 74:361-378. [PMID: 38837539 PMCID: PMC11551836 DOI: 10.1111/pin.13441] [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: 03/09/2024] [Revised: 04/29/2024] [Accepted: 05/12/2024] [Indexed: 06/07/2024]
Abstract
Chronic liver injury induces liver cirrhosis and facilitates hepatocarcinogenesis. However, the effects of this condition on hepatocyte proliferation and differentiation are unclear. We showed that rodent hepatocytes display a ductular phenotype when they are cultured within a collagenous matrix. This process involves transdifferentiation without the emergence of hepatoblastic features and is at least partially reversible. During the ductular reaction in chronic liver diseases with progressive fibrosis, some hepatocytes, especially those adjacent to ectopic ductules, demonstrate ductular transdifferentiation, but the majority of increased ductules originate from the existing bile ductular system that undergoes extensive remodeling. In chronic injury, hepatocyte proliferation is weak but sustained, and most regenerative nodules in liver cirrhosis are composed of clonally proliferating hepatocytes, suggesting that a small fraction of hepatocytes maintain their proliferative capacity in chronic injury. In mouse hepatocarcinogenesis models, hepatocytes activate the expression of various fetal/neonatal genes, indicating that these cells undergo dedifferentiation. Hepatocyte-specific somatic integration of various oncogenes in mice demonstrated that hepatocytes may be the cells of origin for a broad spectrum of liver tumors through transdifferentiation and dedifferentiation. In conclusion, the phenotypic plasticity and heterogeneity of mature hepatocytes are important for understanding the pathogenesis of chronic liver diseases and liver tumors.
Collapse
Affiliation(s)
- Yuji Nishikawa
- President's OfficeAsahikawa Medical UniversityAsahikawaHokkaidoJapan
| |
Collapse
|
3
|
Reddy D, Bhattacharya S, Shah S, Rashid M, Gupta S. DNA methylation mediated downregulation of histone H3 variant H3.3 affects cell proliferation contributing to the development of HCC. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166284. [PMID: 34626773 DOI: 10.1016/j.bbadis.2021.166284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/19/2021] [Accepted: 09/30/2021] [Indexed: 10/20/2022]
Abstract
Chromatin alterations brought by histone variants and modifications potentially regulate gene transcription from tumor initiation to progression. Histone H3.3 variant is one such epigenetic player important for disease progression and development. Though many studies have implicated H3.3 role in cancer progression and metastasis, its regulation, importance of specific modifications and chaperones have been not understood yet. We report DNA methylation mediated downregulation of histone H3 variant H3.3 in HCC and a concomitant increase in the level of the H3.2 variant. The loss of H3.3 in cancer tissues correlates with a decrease in the histone modifications associated with active transcription like H3K9/K14/K27Ac and H3K4Me3. The ectopic overexpression of H3.3 and H3.2 did not affect global PTMs and cell physiology, probably owing to the deregulation of specific histone chaperones CAF-1 (for H3.2) and HIRA (for H3.3) as observed in HCC tissues. Notably, knockdown of P150, a subunit of CAF-1 leads to a cell cycle arrest in S-phase in a neoplastic rat liver cell line, possibly due to the decrease in the histone levels necessary for DNA packaging. Remarkably, modulation of H3.3 in pre-neoplastic rat liver cells lead to an increase in cell proliferation and a decreased transcription of tumor suppressor genes, recapitulating the tumor cell phenotype. Our data suggests, inhibition of DNA methylation and histone deacetylation leads to the restoration of histone H3 variant expression in tumor cells.
Collapse
Affiliation(s)
- Divya Reddy
- Epigenetics and Chromatin Biology Group, Gupta Lab, Cancer Research Institute, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai 410210, MH, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, MH 400085, India.
| | - Saikat Bhattacharya
- Epigenetics and Chromatin Biology Group, Gupta Lab, Cancer Research Institute, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai 410210, MH, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, MH 400085, India.
| | - Sanket Shah
- Epigenetics and Chromatin Biology Group, Gupta Lab, Cancer Research Institute, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai 410210, MH, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, MH 400085, India.
| | - Mudasir Rashid
- Epigenetics and Chromatin Biology Group, Gupta Lab, Cancer Research Institute, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai 410210, MH, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, MH 400085, India.
| | - Sanjay Gupta
- Epigenetics and Chromatin Biology Group, Gupta Lab, Cancer Research Institute, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai 410210, MH, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, MH 400085, India.
| |
Collapse
|
4
|
Abstract
Cardiac hypertrophy, characterized by the enlargement of cardiomyocytes, is initially an adaptive response to physiological and pathological stimuli. Decompensated cardiac hypertrophy is related to fibrosis, inflammatory cytokine, maladaptive remodeling, and heart failure. Although pathological myocardial hypertrophy is the main cause of hypertrophy-related morbidity and mortality, our understanding of its mechanism is still poor. Long noncoding RNAs (lncRNAs) are noncoding RNAs that regulate various physiological and pathological processes through multiple molecular mechanisms. Recently, accumulating evidence has indicated that lncRNA-H19 is a potent regulator of the progression of cardiac hypertrophy. For the first time, this review summarizes the current studies about the role of lncRNA-H19 in cardiac hypertrophy, including its pathophysiological processes and underlying pathological mechanism, including calcium regulation, fibrosis, apoptosis, angiogenesis, inflammation, and methylation. The context within which lncRNA-H19 might be developed as a target for cardiac hypertrophy treatment is then discussed to gain better insight into the possible biological functions of lncRNA-H19 in cardiac hypertrophy.
Collapse
|
5
|
Haddad G, Kölling M, Wegmann UA, Dettling A, Seeger H, Schmitt R, Soerensen-Zender I, Haller H, Kistler AD, Dueck A, Engelhardt S, Thum T, Mueller TF, Wüthrich RP, Lorenzen JM. Renal AAV2-Mediated Overexpression of Long Non-Coding RNA H19 Attenuates Ischemic Acute Kidney Injury Through Sponging of microRNA-30a-5p. J Am Soc Nephrol 2021; 32:323-341. [PMID: 33478972 PMCID: PMC8054899 DOI: 10.1681/asn.2020060775] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 10/28/2020] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Renal ischemia-reperfusion (I/R) injury is a major cause of AKI. Noncoding RNAs are intricately involved in the pathophysiology of this form of AKI. Transcription of hypoxia-induced, long noncoding RNA H19, which shows high embryonic expression and is silenced in adults, is upregulated in renal I/R injury. METHODS Lentivirus-mediated overexpression, as well as antisense oligonucleotide-based silencing, modulated H19 in vitro. In vivo analyses used constitutive H19 knockout mice. In addition, renal vein injection of adeno-associated virus 2 (AAV2) carrying H19 caused overexpression in the kidney. Expression of H19 in kidney transplant patients with I/R injury was investigated. RESULTS H19 is upregulated in kidney biopsies of patients with AKI, in murine ischemic kidney tissue, and in cultured and ex vivo sorted hypoxic endothelial cells (ECs) and tubular epithelial cells (TECs). Transcription factors hypoxia-inducible factor 1-α, LHX8, and SPI1 activate H19 in ECs and TECs. H19 overexpression promotes angiogenesis in vitro and in vivo. In vivo, transient AAV2-mediated H19 overexpression significantly improved kidney function, reduced apoptosis, and reduced inflammation, as well as preserving capillary density and tubular epithelial integrity. Sponging of miR-30a-5p mediated the effects, which, in turn, led to target regulation of Dll4, ATG5, and Snai1. CONCLUSIONS H19 overexpression confers protection against renal injury by stimulating proangiogenic signaling. H19 overexpression may be a promising future therapeutic option in the treatment of patients with ischemic AKI.
Collapse
Affiliation(s)
- George Haddad
- Department of Nephrology, University Hospital Zürich, Zurich, Switzerland
| | - Malte Kölling
- Department of Nephrology, University Hospital Zürich, Zurich, Switzerland
| | - Urs A. Wegmann
- Department of Nephrology, University Hospital Zürich, Zurich, Switzerland
| | - Angela Dettling
- Department of Nephrology, Hannover Medical School, Hannover, Germany
| | - Harald Seeger
- Department of Nephrology, University Hospital Zürich, Zurich, Switzerland
| | - Roland Schmitt
- Department of Nephrology, Hannover Medical School, Hannover, Germany
| | | | - Hermann Haller
- Department of Nephrology, Hannover Medical School, Hannover, Germany
| | - Andreas D. Kistler
- Department of Internal Medicine, Cantonal Hospital Frauenfeld, Frauenfeld, Switzerland
| | - Anne Dueck
- Institute of Pharmacology and Toxicology, Technical University of Munich, Munich, Germany,German Center for Cardiovascular Research, partner site Munich Heart Alliance, Munich, Germany
| | - Stefan Engelhardt
- Institute of Pharmacology and Toxicology, Technical University of Munich, Munich, Germany,German Center for Cardiovascular Research, partner site Munich Heart Alliance, Munich, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany,Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Thomas F. Mueller
- Department of Nephrology, University Hospital Zürich, Zurich, Switzerland
| | - Rudolf P. Wüthrich
- Department of Nephrology, University Hospital Zürich, Zurich, Switzerland
| | - Johan M. Lorenzen
- Department of Nephrology, University Hospital Zürich, Zurich, Switzerland
| |
Collapse
|
6
|
Gamaev L, Mizrahi L, Friehmann T, Rosenberg N, Pappo O, Olam D, Zeira E, Bahar Halpern K, Caruso S, Zucman-Rossi J, Axelrod JH, Galun E, Goldenberg DS. The pro-oncogenic effect of the lncRNA H19 in the development of chronic inflammation-mediated hepatocellular carcinoma. Oncogene 2021; 40:127-139. [PMID: 33093654 DOI: 10.1038/s41388-020-01513-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 10/01/2020] [Accepted: 10/05/2020] [Indexed: 01/09/2023]
Abstract
The oncofetal long noncoding RNA (lncRNA) H19 is postnatally repressed in most tissues, and re-expressed in many cancers, including hepatocellular carcinoma (HCC). The role of H19 in carcinogenesis is a subject of controversy. We aimed to examine the role of H19 in chronic inflammation-mediated hepatocarcinogenesis using the Mdr2/Abcb4 knockout (Mdr2-KO) mouse, a well-established HCC model. For this goal, we have generated Mdr2-KO/H19-KO double knockout (dKO) mice and followed spontaneous tumor development in the dKO and control Mdr2-KO mice. Cellular localization of H19 and effects of H19 loss in the liver were determined in young and old Mdr2-KO mice. Tumor incidence and tumor load were both significantly decreased in the liver of dKO versus Mdr2-KO females. The expression levels of H19 and Igf2 were variable in nontumor liver tissues of Mdr2-KO females and were significantly downregulated in most matched tumors. In nontumor liver tissue of aged Mdr2-KO females, H19 was expressed mainly in hepatocytes, and hepatocyte proliferation was increased compared to dKO females. At an early age, dKO females displayed lower levels of liver injury and B-cell infiltration, with higher percentage of binuclear hepatocytes. In human samples, H19 expression was higher in females, positively correlated with cirrhosis (in nontumor liver samples) and negatively correlated with CTNNB1 (beta-catenin) mutations and patients' survival (in tumors). Our data demonstrate that the lncRNA H19 is pro-oncogenic during the development of chronic inflammation-mediated HCC in the Mdr2-KO mouse model, mainly by increasing liver injury and decreasing hepatocyte polyploidy in young mice.
Collapse
Affiliation(s)
- Lika Gamaev
- The Goldyne Savad Institute of Gene and Cell Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Lina Mizrahi
- The Goldyne Savad Institute of Gene and Cell Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Tomer Friehmann
- The Goldyne Savad Institute of Gene and Cell Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Nofar Rosenberg
- The Goldyne Savad Institute of Gene and Cell Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Orit Pappo
- Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Devorah Olam
- The Goldyne Savad Institute of Gene and Cell Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Evelyne Zeira
- The Goldyne Savad Institute of Gene and Cell Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Keren Bahar Halpern
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Stefano Caruso
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Functional Genomics of Solid Tumors Laboratory, F-75006, Paris, France
| | - Jessica Zucman-Rossi
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Functional Genomics of Solid Tumors Laboratory, F-75006, Paris, France
- Hôpital Européen Georges Pompidou, AP-HP, F-75015, Paris, France
| | - Jonathan H Axelrod
- The Goldyne Savad Institute of Gene and Cell Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Eithan Galun
- The Goldyne Savad Institute of Gene and Cell Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Daniel S Goldenberg
- The Goldyne Savad Institute of Gene and Cell Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel.
| |
Collapse
|
7
|
Zhu Y, Qiu Z, Zhang Y, Li B, Jiang X. Partial hepatectomy‑induced upregulation of SNHG12 promotes hepatocyte proliferation and liver regeneration. Mol Med Rep 2019; 21:1089-1096. [PMID: 31894329 PMCID: PMC7003022 DOI: 10.3892/mmr.2019.10904] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 11/11/2019] [Indexed: 12/13/2022] Open
Abstract
Following partial hepatectomy (PH), the complex process of liver regeneration is initiated, which encompasses the synchronized induction of hepatocyte proliferation. Hepatocyte proliferation can be regulated by multiple stimuli, including long non-coding RNAs (lncRNAs) and Wnt/β-catenin signaling, although the underlying mechanism of lncRNA/Wnt in liver regeneration remains unclear. In the present study, a liver regeneration-associated functional lncRNA was identified, and its function was delineated in vitro and in vivo; lncRNA small nucleolar RNA host gene 12 (SNHG12) was revealed to be upregulated at various time-points after 2/3 PH. The expression of SNHG12 was also increased in normal liver cell lines treated with different concentrations of hepatocyte growth factor (HGF). Functionally, SNHG12 enhanced hepatocyte proliferation in vitro and in vivo, and the liver/body weight ratio of SNHG12-overexpressing mice was significantly higher than that of the control mice. Overexpression of SNHG12 promoted the activation of Wnt/β-catenin signaling in hepatocytes. Furthermore, specific inhibition of Wnt/β-catenin signaling significantly attenuated SNHG12-induced hepatocyte proliferation and the affected liver/body weight ratio. Collectively, the results of the present study indicated that SNHG12 contributes to liver regeneration by activating Wnt/β-catenin signaling. Therefore, drugs that regulate the SNHG12/Wnt axis may be beneficial for liver regeneration following PH.
Collapse
Affiliation(s)
- Yan Zhu
- Department of Pathology, Changhai Hospital, Secondary Military Medicine University, Shanghai 200433, P.R. China
| | - Zhiquan Qiu
- Biliary Tract Surgery Department I, Eastern Hepatobiliary Surgery Hospital, Secondary Military Medicine University, Shanghai 200438, P.R. China
| | - Yiliang Zhang
- Department of Medical Genetics, Secondary Military Medicine University, Shanghai 200433, P.R. China
| | - Bin Li
- Biliary Tract Surgery Department I, Eastern Hepatobiliary Surgery Hospital, Secondary Military Medicine University, Shanghai 200438, P.R. China
| | - Xiaoqing Jiang
- Biliary Tract Surgery Department I, Eastern Hepatobiliary Surgery Hospital, Secondary Military Medicine University, Shanghai 200438, P.R. China
| |
Collapse
|
8
|
Wong DE, Banyard DA, Santos PJF, Sayadi LR, Evans GRD, Widgerow AD. Adipose-derived stem cell extracellular vesicles: A systematic review ✰. J Plast Reconstr Aesthet Surg 2019; 72:1207-1218. [PMID: 30952587 DOI: 10.1016/j.bjps.2019.03.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 02/21/2019] [Accepted: 03/10/2019] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Extracellular vesicles (EVs) are cell-secreted packages that deliver cargo to target cells to effect functional and phenotypic changes. They are secreted by many different cell types, including adipose-derived stem cells (ADSCs), which are a promising field of study in regenerative medicine. Our aim was to perform a systematic review of the literature to summarize the scientific work that has been conducted on ADSC EVs to date. METHODS The Pubmed database was queried with keywords (and variations of) "adipose derived stem cell," "stromal vascular fraction," and "extracellular vesicles." We excluded review papers, then manually screened articles based on title and abstract. Full-text articles were assessed for eligibility to include in final review. RESULTS While an extensive body of research exists on EVs, a much smaller proportion of that is original research on ADSC EVs. Of 44 manuscripts that met our database search criteria, 21 articles were selected for our systematic review. CONCLUSION ADSC EVs were found to exert effects on angiogenesis, cell survival and apoptosis, inflammation, tissue regeneration, and reduction of disease pathology. Further studies examine characteristics of ADSC EVs. Future work should aim to further detail the safety profiles of ADSC EVs given their potential for cell-based therapies. The body of research studies characterizing ADSC EVs continues to expand, and much work remains to be done before human pilot studies can be considered. To our knowledge, we offer the first systematic review summarizing the research on ADSC EVs and their determined roles to date.
Collapse
Affiliation(s)
- Daniel E Wong
- Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Derek A Banyard
- Department of Plastic Surgery, University of California, Irvine, Orange, CA, United States; Center for Tissue Engineering, Department of Plastic Surgery, University of California, Irvine, Orange, CA, United States
| | - Pauline J F Santos
- Department of Plastic Surgery, University of California, Irvine, Orange, CA, United States
| | - Lohrasb R Sayadi
- Center for Tissue Engineering, Department of Plastic Surgery, University of California, Irvine, Orange, CA, United States
| | - Gregory R D Evans
- Department of Plastic Surgery, University of California, Irvine, Orange, CA, United States
| | - Alan D Widgerow
- Department of Plastic Surgery, University of California, Irvine, Orange, CA, United States; Center for Tissue Engineering, Department of Plastic Surgery, University of California, Irvine, Orange, CA, United States.
| |
Collapse
|
9
|
Jiang Y, Huang Y, Cai S, Song Y, Boyer JL, Zhang K, Gao L, Zhao J, Huang W, Liang G, Liangpunsakul S, Wang L. H19 Is Expressed in Hybrid Hepatocyte Nuclear Factor 4α + Periportal Hepatocytes but Not Cytokeratin 19 + Cholangiocytes in Cholestatic Livers. Hepatol Commun 2018; 2:1356-1368. [PMID: 30411082 PMCID: PMC6211330 DOI: 10.1002/hep4.1252] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 07/27/2018] [Indexed: 12/29/2022] Open
Abstract
Long noncoding RNA (lncRNA) H19 is abundantly expressed in fetal liver. Its expression is significantly diminished in adult healthy liver but is re‐induced in chronic liver diseases, including cholestasis. In this study, we developed a new method with combined in situ hybridization (ISH) and immunofluorescence (IF) colabeling to establish an H19 expression profile with both parenchymal and nonparenchymal cell‐specific markers in the livers of cholestatic mouse models and patients with cholestasis. H19RNA+ cells showed no colocalization with biliary epithelial cell marker cytokeratin 19 (CK19)+ cholangiocytes but were immediately adjacent to biliary structures in bile duct ligation (BDL), 3,5‐diethoxycarbony1‐1,4‐dihydrocollidine (DDC), and multidrug‐resistant gene 2 knockout (Mdr2–/–) mouse models and in human primary biliary cholangitis (PBC) and primary sclerosing cholangitis (PSC) liver specimens. In contrast, double‐positive H19RNA+/sex‐determining region Y (SRY)‐box 9 (SOX9)+ ductal progenitor cells, H19RNA+/hepatocyte nuclear factor 4α (HNF4α)+ hepatocytes, H19RNA+/F4/80+ Kupffer cells, HNF4α+/SOX9+ hybrid hepatocytes, as well as triple‐positive H19RNA+/HNF4α+/SOX9+ periportal hepatocytes were identified. In addition, H19RNA could not be detected in mesenchymal cell marker desmin+ cells. Furthermore, H19RNA was predominately detected in cytoplasm with a small amount at the interspace with neighboring cells. Conclusion:H19RNA is localized in HNF4α+ periportal hepatocytes, SOX9+ ductal progenitor cells, and F4/80+ Kupffer cells but not in CK19+ cholangiocytes and desmin+ stellate cells in cholestatic livers.
Collapse
Affiliation(s)
- YanChao Jiang
- Department of Physiology and Neurobiology and the Institute of Systems Genomics University of Connecticut Storrs CT
| | - Yi Huang
- Department of Physiology and Neurobiology and the Institute of Systems Genomics University of Connecticut Storrs CT.,School of Pharmaceutical Sciences Wenzhou Medical University Wenzhou China
| | - ShiYing Cai
- Department of Internal Medicine, Liver Center Yale University New Haven CT
| | - YongFeng Song
- Department of Physiology and Neurobiology and the Institute of Systems Genomics University of Connecticut Storrs CT.,Department of Endocrinology and Metabolism Shandong Provincial Hospital/Shandong University Jinan China
| | - James L Boyer
- Department of Internal Medicine, Liver Center Yale University New Haven CT
| | - KeZhong Zhang
- Center for Molecular Medicine and Genetics Wayne State University School of Medicine Detroit MI
| | - Ling Gao
- Department of Endocrinology and Metabolism Shandong Provincial Hospital/Shandong University Jinan China
| | - JiaJun Zhao
- Department of Endocrinology and Metabolism Shandong Provincial Hospital/Shandong University Jinan China
| | - WenDong Huang
- Department of Diabetes Complications and Metabolism, Diabetes and Metabolism Research Institute Beckman Research Institute, City of Hope National Medical Center Duarte CA
| | - Guang Liang
- School of Pharmaceutical Sciences Wenzhou Medical University Wenzhou China
| | - Suthat Liangpunsakul
- Division of Gastroenterology and Hepatology, Department of Medicine.,Department of Biochemistry and Molecular Biology Indiana University School of Medicine Indianapolis IN.,Roudebush Veterans Affairs Medical Center Indianapolis IN
| | - Li Wang
- Department of Physiology and Neurobiology and the Institute of Systems Genomics University of Connecticut Storrs CT.,Department of Internal Medicine, Liver Center Yale University New Haven CT.,Veterans Affairs Connecticut Healthcare System West Haven CT
| |
Collapse
|
10
|
Jin Y, Wang J, Li H, Gao S, Shi R, Yang D, Wang X, Wang X, Zhu L, Wang X, Chen C, Ning K, Gao Z, Xu J, Fu Q. Extracellular Vesicles Secreted by Human Adipose-derived Stem Cells (hASCs) Improve Survival Rate of Rats with Acute Liver Failure by Releasing lncRNA H19. EBioMedicine 2018; 34:231-242. [PMID: 30077720 PMCID: PMC6116414 DOI: 10.1016/j.ebiom.2018.07.015] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 07/08/2018] [Accepted: 07/12/2018] [Indexed: 02/07/2023] Open
Abstract
It has previously been reported that human adipose-derived stem cells (hASCs) can promote the regeneration of damaged tissues in rats with liver failure through a ‘paracrine effect’. Here we demonstrate a therapeutic effect of hASCs derived Extracellular Vesicles (EVs) on rat models with acute liver failure, as shown by the improvement of the survival rate by >70% compared to controls. Gene sequencing of rat liver revealed an increase in human long-chain non-coding RNA (lncRNA) H19 after hASC-derived EVs transplantation. When the H19 coding sequence was silenced in hASCs and EVs were then collected for treatment of rats with liver failure, we saw a decrease in the survival rate to 40%, compared to treatment with EVs generated from non-silenced hASCs. These data indicate that lncRNA H19 may be a potential therapeutic target for the treatment of liver failure.
Collapse
Affiliation(s)
- Yinpeng Jin
- Shanghai Public Health Clinical Center, Fudan University, Jinshan, Shanghai 201508, PR China.
| | - Junyi Wang
- Shanghai Liver Diseases Research Center, The 85th Hospital of PLA, Shanghai 200235, PR China
| | - Hongchao Li
- Shanghai Liver Diseases Research Center, The 85th Hospital of PLA, Shanghai 200235, PR China
| | - Shane Gao
- East Hospital, Tongji University School of Medicine, Shanghai 200120, PR China
| | - Rongfeng Shi
- Department of Interventional & Vascular Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, PR China
| | - Danjing Yang
- East Hospital, Tongji University School of Medicine, Shanghai 200120, PR China
| | - Xianli Wang
- Institute of Neuroscience and State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, PR China
| | - Xi Wang
- Shanghai Public Health Clinical Center, Fudan University, Jinshan, Shanghai 201508, PR China
| | - Liang Zhu
- East Hospital, Tongji University School of Medicine, Shanghai 200120, PR China
| | - Xiaojin Wang
- Shanghai Liver Diseases Research Center, The 85th Hospital of PLA, Shanghai 200235, PR China
| | - Chengwei Chen
- Shanghai Liver Diseases Research Center, The 85th Hospital of PLA, Shanghai 200235, PR China
| | - Ke Ning
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Zhengliang Gao
- East Hospital, Tongji University School of Medicine, Shanghai 200120, PR China.
| | - Jun Xu
- East Hospital, Tongji University School of Medicine, Shanghai 200120, PR China.
| | - Qingchun Fu
- Shanghai Public Health Clinical Center, Fudan University, Jinshan, Shanghai 201508, PR China.
| |
Collapse
|
11
|
Pope C, Piekos SC, Chen L, Mishra S, Zhong XB. The role of H19, a long non-coding RNA, in mouse liver postnatal maturation. PLoS One 2017; 12:e0187557. [PMID: 29099871 PMCID: PMC5669494 DOI: 10.1371/journal.pone.0187557] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 10/21/2017] [Indexed: 12/16/2022] Open
Abstract
H19 RNA is highly expressed at early postnatal ages and precipitously decreases at a specific time corresponding with increases in expression of genes important for mature liver function, such as drug metabolizing enzymes. H19’s role in the regulation of liver maturation is currently unknown. Using an H19 knockout mouse model to determine the role of H19 in liver development, we quantified gene expression for insulin growth factor signaling, Wnt signaling, key cytochrome P450 (P450) enzymes known to change as the liver develops, and fetal and adult plasma protein produced in liver. In mice lacking H19 expression, liver weights were significantly increased immediately after birth and significant increases were found in the number of actively proliferating cells. Increases in cell proliferation may be due to increases in β-catenin protein affecting Wnt signaling, increases in insulin-like growth factor 2 (IGF2) expression, and/or increases in insulin-like growth factor 1 receptor (IGF1R) expression at the protein level. Loss of targeted inhibition of IGF1R by microRNA 675 (miR-675) may be the cause of IGF1R increases, as miR-675 expression is also abrogated with loss of H19 expression in our model. P450 expression patterns were largely unchanged. No change in the production of plasma proteins was found, indicating H19 may not be important for liver maturation despite its role in controlling cell proliferation during liver growth. H19 may be important for normal liver development, and understanding how the liver matures will assist in predicting drug efficacy and toxicity in pediatric populations.
Collapse
Affiliation(s)
- Chad Pope
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut, United States of America
- * E-mail: ,
| | - Stephanie C. Piekos
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut, United States of America
| | - Liming Chen
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut, United States of America
| | - Shashank Mishra
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut, United States of America
| | - Xiao-bo Zhong
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut, United States of America
| |
Collapse
|
12
|
Pope C, Mishra S, Russell J, Zhou Q, Zhong XB. Targeting H19, an Imprinted Long Non-Coding RNA, in Hepatic Functions and Liver Diseases. Diseases 2017; 5:E11. [PMID: 28933364 PMCID: PMC5456333 DOI: 10.3390/diseases5010011] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 03/03/2017] [Indexed: 12/17/2022] Open
Abstract
H19 is a long non-coding RNA regulated by genomic imprinting through methylation at the locus between H19 and IGF2. H19 is important in normal liver development, controlling proliferation and impacting genes involved in an important network controlling fetal development. H19 also plays a major role in disease progression, particularly in hepatocellular carcinoma. H19 participates in the epigenetic regulation of many processes impacting diseases, such as activating the miR-200 pathway by histone acetylation to inhibit the epithelial-mesenchymal transition to suppress tumor metastasis. Furthermore, H19's normal regulation is disturbed in diseases, such as hepatocellular carcinoma. In this disease, aberrant epigenetic maintenance results in biallelic expression of IGF2, leading to uncontrolled cellular proliferation. This review aims to further research utilizing H19 for drug discovery and the treatment of liver diseases by focusing on both the epigenetic regulation of H19 and how H19 regulates normal liver functions and diseases, particularly by epigenetic mechanisms.
Collapse
Affiliation(s)
- Chad Pope
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, 69 N Eagleville Road, Storrs, CT 06269, USA.
| | - Shashank Mishra
- Department of Physiology and Neurobiology, University of Connecticut, 75 N Eagleville Road, Storrs, CT 06269, USA.
| | - Joshua Russell
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, 69 N Eagleville Road, Storrs, CT 06269, USA.
| | - Qingqing Zhou
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, 69 N Eagleville Road, Storrs, CT 06269, USA.
| | - Xiao-Bo Zhong
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, 69 N Eagleville Road, Storrs, CT 06269, USA.
| |
Collapse
|
13
|
Li C, Chang L, Chen Z, Liu Z, Wang Y, Ye Q. The role of lncRNA MALAT1 in the regulation of hepatocyte proliferation during liver regeneration. Int J Mol Med 2017; 39:347-356. [PMID: 28075444 PMCID: PMC5358694 DOI: 10.3892/ijmm.2017.2854] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Accepted: 12/29/2016] [Indexed: 12/22/2022] Open
Abstract
Exploring the biological functions of long non-coding RNAs (lncRNAs) has come to the foreground in recent years. Studies have indicated that the lncRNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) not only regulates tumorigenesis in hepatocellular carcinoma, but also controls cell cycle progression in hematopoietic cells. The present study was designed to investigate the biological role of lncRNA MALAT1 in liver regeneration. We carried out a series of assays during liver regeneration following 2/3 partial hepatectomy in mice. We explored the functions of lncRNA MALAT1 with a series of functional analyses in vitro. We found that MALAT1 was upregulated during liver regeneration. Moreover, MALAT1 accelerated hepatocyte proliferation by stimulating cell cycle progression from the G1 to the S phase and inhibiting apoptosis in vitro. In addition, our findings also demonstrated that MALAT1 was regulated by p53 during liver regeneration, and that p53 may be a key upstream regulator of MALAT1 activity. Mechanistically, we found that MALAT1 activated the Wnt/β-catenin pathway by inhibiting the expression of Axin1 and adenomatous polyposis coli (APC), and subsequently promoting the expression of cyclin D1. On the whole, the findings of this study suggest that MALAT1 is a critical molecule for liver regeneration. Pharmacological interventions targeting MALAT1 may thus prove to be therapeutically beneficial in liver failure or liver transplantation by promoting liver regeneration.
Collapse
Affiliation(s)
- Cuicui Li
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, Hubei 430071, P.R. China
| | - Lei Chang
- Department of General Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Zhiquan Chen
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, Hubei 430071, P.R. China
| | - Zhongzhong Liu
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, Hubei 430071, P.R. China
| | - Yanfeng Wang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, Hubei 430071, P.R. China
| | - Qifa Ye
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
14
|
Ramani K, Mavila N, Ko KS, Mato JM, Lu SC. Prohibitin 1 Regulates the H19-Igf2 Axis and Proliferation in Hepatocytes. J Biol Chem 2016; 291:24148-24159. [PMID: 27687727 DOI: 10.1074/jbc.m116.744045] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 09/26/2016] [Indexed: 12/15/2022] Open
Abstract
Prohibitin 1 (PHB1) is a mitochondrial chaperone that regulates cell growth. Phb1 knock-out mice exhibit liver injury and hepatocellular carcinoma (HCC). Phb1 knock-out livers show induction of tumor growth-associated genes, H19 and insulin-like growth factor 2 (Igf2). These genes are controlled by the imprinting control region (ICR) containing CCCTC-binding transcription factor (CTCF)-binding sites. Because Phb1 knock-out mice exhibited induction of H19 and Igf2, we hypothesized that PHB1-mediated regulation of the H19-Igf2 axis might control cell proliferation in normal hepatocytes. H19 and Igf2 were induced (8-20-fold) in 3-week-old Phb1 knock-out livers, in Phb1 siRNA-treated AML12 hepatocytes (2-fold), and HCC cell lines when compared with control. Phb1 knockdown lowered CTCF protein in AML12 by ∼30% when compared with control. CTCF overexpression lowered basal H19 and Igf2 expression by 30% and suppressed Phb1 knockdown-mediated induction of these genes. CTCF and PHB1 co-immunoprecipitated and co-localized on the ICR element, and Phb1 knockdown lowered CTCF ICR binding activity. The results suggest that PHB1 and CTCF cooperation may control the H19-Igf2 axis. Human HCC tissues with high levels of H19 and IGF2 exhibited a 40-50% reduction in PHB1 and CTCF expression and their ICR binding activity. Silencing Phb1 or overexpressing H19 in the mouse HCC cell line, SAMe-D, induced cell growth. Blocking H19 induction prevented Phb1 knockdown-mediated growth, whereas H19 overexpression had the reverse effect. Interestingly H19 silencing induced PHB1 expression. Taken together, our results demonstrate that the H19-Igf2 axis is negatively regulated by CTCF-PHB1 cooperation and that H19 is involved in modulating the growth-suppressive effect of PHB1 in the liver.
Collapse
Affiliation(s)
- Komal Ramani
- From the Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, California 90048
| | - Nirmala Mavila
- From the Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, California 90048
| | - Kwang Suk Ko
- the Department of Nutritional Science and Food Management, the College of Health Science, Ewha Womans University, Seoul 03760, Korea, and
| | - José M Mato
- the CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain
| | - Shelly C Lu
- From the Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, California 90048,
| |
Collapse
|
15
|
Boutin SR, Rogers AB, Shen Z, Fry RC, Love JA, Nambiar PR, Suerbaum S, Fox JG. Hepatic Temporal Gene Expression Profiling in Helicobacter hepaticus-Infected A/JCr Mice. Toxicol Pathol 2016; 32:678-93. [PMID: 15513911 DOI: 10.1080/01926230490524058] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Helicobacter hepaticus infection of A/JCr mice is a model of infectious liver cancer. We monitored hepatic global gene expression profiles in H. hepaticus infected and control male A/JCr mice at 3 months, 6 months, and 1 year of age using an Affymetrix-based oligonucleotide microarray platform on the premise that a specific genetic expression signature at isolated time points would be indicative of disease status. Model based expression index comparisons generated by dChip yielded consistent profiles of differential gene expression for H. hepaticus infected male mice with progressive liver disease versus uninfected control mice within each age group. Linear discriminant analysis and principal component analysis allowed segregation of mice based on combined age and lesion status, or age alone. Up-regulation of putative tumor markers correlated with advancing hepatocellular dysplasia. Transcriptionally down-regulated genes in mice with liver lesions included those related to peroxisome proliferator, fatty acid, and steroid metabolism pathways. In conclusion, transcriptional profiling of hepatic genes documented gene expression signatures in the livers of H. hepaticus infected male A/JCr mice with chronic progressive hepatitis and preneoplastic liver lesions, complemented the histopathological diagnosis, and suggested molecular targets for the monitoring and intervention of disease progression prior to the onset of hepatocellular neoplasia.
Collapse
Affiliation(s)
- Samuel R Boutin
- Division of Comparative Medicine, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, USA
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Konishi H, Ichikawa D, Yamamoto Y, Arita T, Shoda K, Hiramoto H, Hamada J, Itoh H, Fujita Y, Komatsu S, Shiozaki A, Ikoma H, Ochiai T, Otsuji E. Plasma level of metastasis-associated lung adenocarcinoma transcript 1 is associated with liver damage and predicts development of hepatocellular carcinoma. Cancer Sci 2016; 107:149-54. [PMID: 26614531 PMCID: PMC4768388 DOI: 10.1111/cas.12854] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 11/16/2015] [Accepted: 11/20/2015] [Indexed: 12/26/2022] Open
Abstract
Recent studies have shown that metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) was overexpressed in many human solid cancers, however, its roles in plasma of hepatocellular carcinoma (HCC) patients were unclear. The aim of this study was to investigate the significance of plasma MALAT1 levels in HCC patients. Plasma samples were collected from pre-operative HCC, hepatic disease patients, and healthy controls, and tissue samples from HCC patients and colorectal cancer patients with liver metastasis. Plasma and tissue MALAT1 levels were measured. Plasma MALAT1 levels were progressively and significantly higher in HCC patients than hepatic disease patients, and higher in hepatic disease patients than healthy controls. The expression of MALAT1 in HCC tissue was slightly higher than that in paired non-cancerous liver tissue, but not significant. The expression of MALAT1 in the non-cancerous liver tissue of 20 HCC patients was significantly higher than that in normal liver tissue of 13 colorectal cancer patients. In contrast, plasma MALAT1 levels were significantly low in HCC patients with hepatitis B infection, and significantly high in patients with liver damage B or liver cirrhosis. In a receiver-operator curve analysis of HCC and hepatic disease patients, the cut-off value of plasma MALAT1 was 1.60 and the area under the curve was 0.66. Plasma MALAT1 levels were not correlated with α-fetoprotein or protein induced by vitamin K absence II, whereas sensitivity and specificity for the detection of HCC with the combination of MALAT1, α-fetoprotein, and protein induced by vitamin K absence II were 88.6% and 75%, respectively. In conclusion, the plasma MALAT1 level is associated with liver damage, and has clinical utility for predicting development of HCC.
Collapse
Affiliation(s)
- Hirotaka Konishi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Daisuke Ichikawa
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yusuke Yamamoto
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tomohiro Arita
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Katsutoshi Shoda
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hidekazu Hiramoto
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Junichi Hamada
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroshi Itoh
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yuji Fujita
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shuhei Komatsu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Atsushi Shiozaki
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hisashi Ikoma
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Toshiya Ochiai
- Department of Surgery, North Medical Center, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Eigo Otsuji
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
17
|
Chen X, Yamamoto M, Fujii K, Nagahama Y, Ooshio T, Xin B, Okada Y, Furukawa H, Nishikawa Y. Differential reactivation of fetal/neonatal genes in mouse liver tumors induced in cirrhotic and non-cirrhotic conditions. Cancer Sci 2015; 106:972-81. [PMID: 26011625 PMCID: PMC4556385 DOI: 10.1111/cas.12700] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 05/06/2015] [Accepted: 05/17/2015] [Indexed: 12/13/2022] Open
Abstract
Hepatocellular carcinoma develops in either chronically injured or seemingly intact livers. To explore the tumorigenic mechanisms underlying these different conditions, we compared the mRNA expression profiles of mouse hepatocellular tumors induced by the repeated injection of CCl4 or a single diethylnitrosamine (DEN) injection using a cDNA microarray. We identified tumor-associated genes that were expressed differentially in the cirrhotic CCl4 model (H19, Igf2, Cbr3, and Krt20) and the non-cirrhotic DEN model (Tff3, Akr1c18, Gpc3, Afp, and Abcd2) as well as genes that were expressed comparably in both models (Ly6d, Slpi, Spink3, Scd2, and Cpe). The levels and patterns of mRNA expression of these genes were validated by quantitative RT-PCR analyses. Most of these genes were highly expressed in mouse livers during the fetal/neonatal periods. We also examined the mRNA expression of these genes in mouse tumors induced by thioacetamide, another cirrhotic inducer, and those that developed spontaneously in non-cirrhotic livers and found that they shared a similar expression profile as that observed in CCl4-induced and DEN-induced tumors, respectively. There was a close relationship between the expression levels of Igf2 and H19 mRNA, which were activated in the cirrhotic models. Our results show that mouse liver tumors reactivate fetal/neonatal genes, some of which are specific to cirrhotic or non-cirrhotic modes of pathogenesis.
Collapse
Affiliation(s)
- Xi Chen
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Masahiro Yamamoto
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Kiyonaga Fujii
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Yasuharu Nagahama
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Takako Ooshio
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Bing Xin
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Yoko Okada
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Hiroyuki Furukawa
- Division of Gastroenterological and General Surgery, Department of Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Yuji Nishikawa
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| |
Collapse
|
18
|
Xu D, Yang F, Yuan JH, Zhang L, Bi HS, Zhou CC, Liu F, Wang F, Sun SH. Long noncoding RNAs associated with liver regeneration 1 accelerates hepatocyte proliferation during liver regeneration by activating Wnt/β-catenin signaling. Hepatology 2013; 58:739-51. [PMID: 23483581 DOI: 10.1002/hep.26361] [Citation(s) in RCA: 152] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Accepted: 02/20/2013] [Indexed: 12/19/2022]
Abstract
UNLABELLED In recent years, long noncoding RNAs (lncRNAs) have been investigated as a new class of regulators of biological function. A recent study reported that lncRNAs control cell proliferation in hepatocellular carcinoma (HCC). However, the role of lncRNAs in liver regeneration and the overall mechanisms remain largely unknown. To address this issue, we carried out a genome-wide lncRNA microarray analysis during liver regeneration in mice after 2/3 partial hepatectomy (PH) at various timepoints. The results revealed differential expression of a subset of lncRNAs, notably a specific differentially expressed lncRNA associated with Wnt/β-catenin signaling during liver regeneration (an lncRNA associated with liver regeneration, termed lncRNA-LALR1). The functions of lncRNA-LALR1 were assessed by silencing and overexpressing this lncRNA in vitro and in vivo. We found that lncRNA-LALR1 enhanced hepatocyte proliferation by promoting progression of the cell cycle in vitro. Furthermore, we showed that lncRNA-LALR1 accelerated mouse hepatocyte proliferation and cell cycle progression during liver regeneration in vivo. Mechanistically, we discovered that lncRNA-LALR1 facilitated cyclin D1 expression through activation of Wnt/β-catenin signaling by way of suppression of Axin1. In addition, lncRNA-LALR1 inhibited the expression of Axin1 mainly by recruiting CTCF to the AXIN1 promoter region. We also identified a human ortholog RNA of lncRNA-LALR1 (lncRNA-hLALR1) and found that it was expressed in human liver tissues. CONCLUSION lncRNA-LALR1 promotes cell cycle progression and accelerates hepatocyte proliferation during liver regeneration by activating Wnt/β-catenin signaling. Pharmacological intervention targeting lncRNA-LALR1 may be therapeutically beneficial in liver failure and liver transplantation by inducing liver regeneration.
Collapse
Affiliation(s)
- Dan Xu
- Department of Medical Genetics, Second Military Medical University, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Kogure T, Costinean S, Yan I, Braconi C, Croce C, Patel T. Hepatic miR-29ab1 expression modulates chronic hepatic injury. J Cell Mol Med 2012; 16:2647-54. [PMID: 22469499 PMCID: PMC3923513 DOI: 10.1111/j.1582-4934.2012.01578.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Accepted: 03/27/2012] [Indexed: 12/31/2022] Open
Abstract
MicroRNAs (miRNAs) are small, regulatory non-coding RNAs that have potent effects on gene expression. Several miRNA are deregulated in cellular processes involved in human liver diseases and regulation of cellular processes. Recent studies have identified the involvement of miR-29 in hepatic fibrosis and carcinogenesis. Although several targets of miR-29 have been identified, there is limited information regarding the cell-type specific roles of miR-29 in the liver, and we sought to evaluate the role of this miRNA in hepatic pathobiology. We report the generation of a tissue-specific knockout mouse to evaluate the role of miR-29 in hepatic fibrosis and carcinogenesis in response to injury. We hypothesized that miR-29 contributes to the hepatocyte driven response to chronic cellular injury that results in fibrosis. In support of this hypothesis, fibrosis and mortality were enhanced in miR29 knockout mice in response to carbon tetrachloride. Genome-wide gene expression analysis identified an over-representation of genes associated with fibrosis. The oncofetal RNA H19 was modulated in a miR-29 dependent manner following exposure to carbon tetrachloride in vivo. The impact of a hepatocyte specific miR-29 knockout on survival following chronic hepatic injury in vivo implicates this miRNA as a potential target for intervention. These results provide evidence of the involvement of miR-29 in chronic hepatic injury, and suggest a role for deregulated hepatocyte expression of miR-29 in the response to hepatic injury, fibrosis and carcinogenesis.
Collapse
Affiliation(s)
- Takayuki Kogure
- College of Medicine, Ohio State UniversityColumbus, OH, USA
- Department of Transplantation, Division of Gastroenterology and Hepatology, Mayo ClinicJacksonville, FL, USA
| | | | - Irene Yan
- Department of Transplantation, Division of Gastroenterology and Hepatology, Mayo ClinicJacksonville, FL, USA
| | - Chiara Braconi
- College of Medicine, Ohio State UniversityColumbus, OH, USA
| | - Carlo Croce
- College of Medicine, Ohio State UniversityColumbus, OH, USA
| | - Tushar Patel
- College of Medicine, Ohio State UniversityColumbus, OH, USA
- Department of Transplantation, Division of Gastroenterology and Hepatology, Mayo ClinicJacksonville, FL, USA
| |
Collapse
|
20
|
Zhang Q, Pu R, Du Y, Han Y, Su T, Wang H, Cao G. Non-coding RNAs in hepatitis B or C-associated hepatocellular carcinoma: potential diagnostic and prognostic markers and therapeutic targets. Cancer Lett 2012; 321:1-12. [PMID: 22425745 DOI: 10.1016/j.canlet.2012.03.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2012] [Revised: 03/07/2012] [Accepted: 03/08/2012] [Indexed: 02/06/2023]
Abstract
Non-coding RNA (ncRNA), a class of RNAs that do not code protein but have regulatory functions, can regulate gene expression and replication of hepatitis B virus or hepatitis C virus and play an important role in the virus-host interaction and the development of hepatocellular carcinoma (HCC). Deregulated ncRNAs in surgically removed hepatic tissues and circulation can be prognostic and diagnostic markers, respectively. ncRNAs functioning as either tumor suppressors or oncogenes can be therapeutic options. Here, we summarize the deregulated ncRNAs associated with the infections and HCC and focus on their roles on early diagnosis, prognosis prediction and therapeutic option of HCC.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Epidemiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
21
|
Teramura T, Onodera Y, Murakami H, Ito S, Mihara T, Takehara T, Kato H, Mitani T, Anzai M, Matsumoto K, Saeki K, Fukuda K, Sagawa N, Osoi Y. Mouse androgenetic embryonic stem cells differentiated to multiple cell lineages in three embryonic germ layers in vitro. J Reprod Dev 2009; 55:283-92. [PMID: 19305126 DOI: 10.1262/jrd.20146] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The embryos of some rodents and primates can precede early development without the process of fertilization; however, they cease to develop after implantation because of restricted expressions of imprinting genes. Asexually developed embryos are classified into parthenote/gynogenote and androgenote by their genomic origins. Embryonic stem cells (ESCs) derived from asexual origins have also been reported. To date, ESCs derived from parthenogenetic embryos (PgESCs) have been established in some species, including humans, and the possibility to be alternative sources for autologous cell transplantation in regenerative medicine has been proposed. However, some developmental characteristics, which might be important for therapeutic applications, such as multiple differentiation capacity and transplantability of the ESCs of androgenetic origin (AgESCs) are uncertain. Here, we induced differentiation of mouse AgESCs and observed derivation of neural cells, cardiomyocytes and hepatocytes in vitro. Following differentiated embryoid body (EB) transplantation in various mouse strains including the strain of origin, we found that the EBs could engraft in theoretically MHC-matched strains. Our results indicate that AgESCs possess at least two important characteristics, multiple differentiation properties in vitro and transplantability after differentiation, and suggest that they can also serve as a source of histocompatible tissues for transplantation.
Collapse
Affiliation(s)
- Takeshi Teramura
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Mie University.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Erin N, Wang N, Xin P, Bui V, Weisz J, Barkan GA, Zhao W, Shearer D, Clawson GA. Altered gene expression in breast cancer liver metastases. Int J Cancer 2009; 124:1503-16. [PMID: 19117052 DOI: 10.1002/ijc.24131] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
We previously developed a highly aggressive cell line from heart metastases of 4T1 breast carcinoma (designated 4THM), which produced liver metastases (designated 4TLM). In this study, gene array analysis (GAEA) compared gene expression profiles in 4TLM with profiles in 4T1 and 4THM primary tumors. GAEA demonstrated that 4T1 and 4THM tumors differed in about 250 genes. Over 1,000 genes, however, were expressed differently in 4TLM compared with primary tumors. A cohort of 16 genes showed significantly decreased expression in 4THM tumors, which decreased even further in 4TLM. Many of these genes have been implicated in breast cancer, and many are involved in cell adhesion and junctional complexes. Expression of multiple tight and adherence junction proteins was either downregulated or disappeared in 4TLM; downregulation of claudin 4, claudin 7 and gamma-catenin was confirmed by quantitative polymerase chain reaction, immunoblot, and immunocytochemical (ICC) analyses. At the protein level, intact ZO-1 was also observed in 4T1 tumors, but was not expressed in 4THM or 4TLM tumors. ICC demonstrated expression of gamma-catenin at the plasma membrane with 4T1 tumors, whereas staining appeared to be nuclear/perinuclear in 4THM tumors. Claudin 7 staining was also seen in monocyte/pmacrophage-like cells in liver around metastatic lesions by ICC, and it appeared that larger 4TLM tumors apparently reexpressed claudin 7 RNA and protein. Our results demonstrate that decreased or abnormal expression of a number of cell adhesion/junctional proteins, including claudin 4, 7, ZO-1 and gamma-catenin, correlates with liver metastases, and that cell adhesion molecules in the microenvironment are also altered.
Collapse
Affiliation(s)
- Nuray Erin
- Department of Pathology, Gittlen Cancer Research Foundation, Hershey Medical Center, Pennsylvania State University, Hershey, PA 17033, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Khatib H, Schutzkus V. The expression profile of the H19 gene in cattle. Mamm Genome 2006; 17:991-6. [PMID: 16964441 DOI: 10.1007/s00335-006-0038-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2006] [Accepted: 04/25/2006] [Indexed: 12/01/2022]
Abstract
The expression of the H19 gene has been well studied in fetal human and mouse tissues but not in cattle. It is generally believed that H19 is abundantly expressed in the early stages of embryogenesis and is repressed postnatally. We report the expression pattern of this gene in cattle in a total of 120 fetal-organ combinations and in a total of 108 adult-organ combinations using quantitative real time PCR. In fetal tissues, H19 was abundantly expressed in amnion, chorion, and allantois. Fetal liver, lung, heart, spleen, eye, rib, mammary gland, and cotyledon showed moderate expression, while intestine and brain showed lower expression levels. For some organs examined in this study, the expression pattern in cattle fetal organs was similar to that of human, mouse, and sheep. For adult organs, H19 was highly expressed in muscle and moderately expressed in liver, lung, heart, kidney, pancreas, and ovary. Low expression levels were observed for adult spleen, caruncle, and endometrium. Of considerable interest was the observation that H19 transcripts have not been detected in the mouse and human kidney or in the mouse spleen. In contrast, we observed significant expression levels in adult kidney and low expression levels in adult spleen. In a comparison of adults and fetuses, significant differences in H19 expression levels were found for liver, lung, heart, and spleen. The expression pattern in adults implies that, in addition to possible roles in embryogenesis, the H19 gene may have other functions.
Collapse
Affiliation(s)
- Hasan Khatib
- Department of Dairy Science, University of Wisconsin-Madison, 1675 Observatory Drive, Madison, WI 53706, USA.
| | | |
Collapse
|
24
|
Piscaglia AC, Zocco MA, Di Campli C, Sparano L, Rutella S, Monego G, Bonanno G, Michetti F, Mancuso S, Pola P, Leone G, Gasbarrini G, Gasbarrini A. How does human stem cell therapy influence gene expression after liver injury? Microarray evaluation on a rat model. Dig Liver Dis 2005; 37:952-963. [PMID: 16214431 DOI: 10.1016/j.dld.2005.06.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2005] [Accepted: 06/22/2005] [Indexed: 12/11/2022]
Abstract
BACKGROUND Tissue homeostasis is guaranteed by stem proliferating reserve, depending on dynamic changes in gene expression. A high plasticity is shown by the haematopoietic stem cells, potential source for liver regeneration. AIM We aimed to evaluate the gene expression modifications induced by human haematopoietic stem cell therapy after liver injury in rats. SUBJECTS Rats were sorted as follows: (A) human-haematopoietic stem cell injection after allyl alcohol liver damage; (B) only haematopoietic stem cell injection; (C) only allyl alcohol injection; and (D) sacrifice without any treatment. METHODS Livers, spleens and bone marrows were analysed with flow-cytometry. Livers were also studied by reverse-transcription PCR, histology, immunohistochemistry and microarray analysis; selected genes were confirmed by real-time PCR. RESULTS In subset A, haematopoietic stem cells were selectively recruited by liver, with respect to the group B, and they improved the liver regeneration process compared to group C. As regards microarrays, haematopoietic stem cell infusion upregulates 265 genes and downregulates 149 genes. Differentially regulated genes belong to a broad range of functional pathways, including proliferation, differentiation, adhesion/migration and transcripts related to oval-cell activation. Real-time PCR validated array results. CONCLUSIONS Our study confirmed the capacity of haematopoietic stem cells to contribute to liver regeneration. Moreover, microarray analysis led to the identification of genes whose regulation strongly correlates with a more efficient process of liver repair after haematopoietic stem cell injection.
Collapse
Affiliation(s)
- A C Piscaglia
- Department of Internal Medicine and Gastroenterology, Catholic University of Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|