51
|
Elsegood CL, Chan CW, Degli-Esposti MA, Wikstrom ME, Domenichini A, Lazarus K, van Rooijen N, Ganss R, Olynyk JK, Yeoh GCT. Kupffer cell-monocyte communication is essential for initiating murine liver progenitor cell-mediated liver regeneration. Hepatology 2015; 62:1272-84. [PMID: 26173184 DOI: 10.1002/hep.27977] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 06/10/2015] [Accepted: 07/04/2015] [Indexed: 12/27/2022]
Abstract
UNLABELLED Liver progenitor cells (LPCs) are necessary for repair in chronic liver disease because the remaining hepatocytes cannot replicate. However, LPC numbers also correlate with disease severity and hepatocellular carcinoma risk. Thus, the progenitor cell response in diseased liver may be regulated to optimize liver regeneration and minimize the likelihood of tumorigenesis. How this is achieved is currently unknown. Human and mouse diseased liver contain two subpopulations of macrophages with different ontogenetic origins: prenatal yolk sac-derived Kupffer cells and peripheral blood monocyte-derived macrophages. We examined the individual role(s) of Kupffer cells and monocyte-derived macrophages in the induction of LPC proliferation using clodronate liposome deletion of Kupffer cells and adoptive transfer of monocytes, respectively, in the choline-deficient, ethionine-supplemented diet model of liver injury and regeneration. Clodronate liposome treatment reduced initial liver monocyte numbers together with the induction of injury and LPC proliferation. Adoptive transfer of monocytes increased the induction of liver injury, LPC proliferation, and tumor necrosis factor-α production. CONCLUSION Kupffer cells control the initial accumulation of monocyte-derived macrophages. These infiltrating monocytes are in turn responsible for the induction of liver injury, the increase in tumor necrosis factor-α, and the subsequent proliferation of LPCs.
Collapse
Affiliation(s)
- Caryn L Elsegood
- School of Chemistry and Biochemistry, The University of Western Australia, Crawley, Western Australia, Australia.,School of Biomedical Sciences, CHIRI Biosciences Research Precinct, Curtin University, Bentley, Western Australia, Australia
| | - Chun Wei Chan
- School of Medicine and Pharmacology, The University of Western Australia, Fremantle, Western Australia, Australia.,School of Biological Sciences and Biotechnology, Murdoch University, Murdoch, Western Australia, Australia
| | - Mariapia A Degli-Esposti
- Immunology and Virology Program, Centre for Ophthalmology and Visual Science, The University of Western Australia, Nedlands, Western Australia, Australia.,Centre for Experimental Immunology, Lions Eye Institute, Nedlands, Western Australia, Australia
| | - Matthew E Wikstrom
- Immunology and Virology Program, Centre for Ophthalmology and Visual Science, The University of Western Australia, Nedlands, Western Australia, Australia.,Centre for Experimental Immunology, Lions Eye Institute, Nedlands, Western Australia, Australia
| | - Alice Domenichini
- School of Biomedical Sciences, CHIRI Biosciences Research Precinct, Curtin University, Bentley, Western Australia, Australia
| | - Kyren Lazarus
- School of Biomedical Sciences, CHIRI Biosciences Research Precinct, Curtin University, Bentley, Western Australia, Australia.,Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Crawley, Western Australia, Australia
| | - Nico van Rooijen
- Department of Molecular Cell Biology, VU Medical Center, Amsterdam, The Netherlands
| | - Ruth Ganss
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Crawley, Western Australia, Australia
| | - John K Olynyk
- School of Biomedical Sciences, CHIRI Biosciences Research Precinct, Curtin University, Bentley, Western Australia, Australia.,Department of Gastroenterology and Hepatology, Fiona Stanley and Fremantle Hospitals, South Metropolitan Health Service, Western Australia, Australia.,Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, Australia
| | - George C T Yeoh
- School of Chemistry and Biochemistry, The University of Western Australia, Crawley, Western Australia, Australia.,Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Crawley, Western Australia, Australia
| |
Collapse
|
52
|
Abstract
In recent years, hepatic oval cells (HOC) have gradually become a research hotspot, and their participation in the reconstruction of liver structure and function has been preliminarily confirmed. This provides a new direction for the study of the pathogenesis and treatment of liver injury, hepatitis, liver fibrosis, cirrhosis, liver neoplasms and other liver diseases. This paper will discuss the relationship between hepatic oval cells and liver diseases.
Collapse
|
53
|
Becker RA, Patlewicz G, Simon TW, Rowlands JC, Budinsky RA. The adverse outcome pathway for rodent liver tumor promotion by sustained activation of the aryl hydrocarbon receptor. Regul Toxicol Pharmacol 2015; 73:172-90. [PMID: 26145830 DOI: 10.1016/j.yrtph.2015.06.015] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 06/19/2015] [Accepted: 06/22/2015] [Indexed: 12/29/2022]
Abstract
An Adverse Outcome Pathway (AOP) represents the existing knowledge of a biological pathway leading from initial molecular interactions of a toxicant and progressing through a series of key events (KEs), culminating with an apical adverse outcome (AO) that has to be of regulatory relevance. An AOP based on the mode of action (MOA) of rodent liver tumor promotion by dioxin-like compounds (DLCs) has been developed and the weight of evidence (WoE) of key event relationships (KERs) evaluated using evolved Bradford Hill considerations. Dioxins and DLCs are potent aryl hydrocarbon receptor (AHR) ligands that cause a range of species-specific adverse outcomes. The occurrence of KEs is necessary for inducing downstream biological responses and KEs may occur at the molecular, cellular, tissue and organ levels. The common convention is that an AOP begins with the toxicant interaction with a biological response element; for this AOP, this initial event is binding of a DLC ligand to the AHR. Data from mechanistic studies, lifetime bioassays and approximately thirty initiation-promotion studies have established dioxin and DLCs as rat liver tumor promoters. Such studies clearly show that sustained AHR activation, weeks or months in duration, is necessary to induce rodent liver tumor promotion--hence, sustained AHR activation is deemed the molecular initiating event (MIE). After this MIE, subsequent KEs are 1) changes in cellular growth homeostasis likely associated with expression changes in a number of genes and observed as development of hepatic foci and decreases in apoptosis within foci; 2) extensive liver toxicity observed as the constellation of effects called toxic hepatopathy; 3) cellular proliferation and hyperplasia in several hepatic cell types. This progression of KEs culminates in the AO, the development of hepatocellular adenomas and carcinomas and cholangiolar carcinomas. A rich data set provides both qualitative and quantitative knowledge of the progression of this AOP through KEs and the KERs. Thus, the WoE for this AOP is judged to be strong. Species-specific effects of dioxins and DLCs are well known--humans are less responsive than rodents and rodent species differ in sensitivity between strains. Consequently, application of this AOP to evaluate potential human health risks must take these differences into account.
Collapse
Affiliation(s)
- Richard A Becker
- Regulatory and Technical Affairs Department, American Chemistry Council (ACC), Washington, DC 20002, USA.
| | - Grace Patlewicz
- DuPont Haskell Global Centers for Health and Environmental Sciences, Newark, DE 19711, USA
| | - Ted W Simon
- Ted Simon LLC, 4184 Johnston Road, Winston, GA 30187, USA
| | - J Craig Rowlands
- The Dow Chemical Company, Toxicology & Environmental Research & Consulting, 1803 Building Washington Street, Midland, MI 48674, USA
| | - Robert A Budinsky
- The Dow Chemical Company, Toxicology & Environmental Research & Consulting, 1803 Building Washington Street, Midland, MI 48674, USA
| |
Collapse
|
54
|
Chen J, Zhang X, Xu Y, Li X, Ren S, Zhou Y, Duan Y, Zern M, Zhang H, Chen G, Liu C, Mu Y, Liu P. Hepatic Progenitor Cells Contribute to the Progression of 2-Acetylaminofluorene/Carbon Tetrachloride-Induced Cirrhosis via the Non-Canonical Wnt Pathway. PLoS One 2015; 10:e0130310. [PMID: 26087010 PMCID: PMC4473299 DOI: 10.1371/journal.pone.0130310] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 05/19/2015] [Indexed: 12/16/2022] Open
Abstract
Hepatic progenitor cells (HPCs) appear to play an important role in chronic liver injury. In this study, cirrhosis was induced in F-344 rats (n = 32) via subcutaneous injection of 50% carbon tetrachloride (CCl4) twice a week for 8 weeks. Then, 30% CCl4 was administered in conjunction with intragastric 2-acetylaminofluorine (2-AAF) for 4 weeks to induce activation of HPCs. WB-F344 cells were used to provide direct evidence for differentiation of HPCs to myofibroblasts. The results showed that after administration of 2-AAF, the hydroxyproline content and the expressions of α-SMA, Col I, Col IV, TGF-β1, CD68, TNF-α, CK19 and OV6 were significantly increased. OV6 and α-SMA were largely co-expressed in fibrous septum and the expressions of Wnt5b, frizzled2, frizzled3 and frizzled6 were markedly increased, while β-catenin expression was not statistically different among the different groups. Consistent with the above results, WB-F344 cells, treated with TGF-β1 in vitro, differentiated into myofibroblasts and α-SMA, Col I, Col IV, Wnt5b and frizzled2 expressions were significantly increased, while β-catenin expression was decreased. After blocking the non-canonical Wnt pathway via WIF-1, the Wnt5b level was down regulated, and α-SMA and F-actin expressions were significantly decreased in the WIF-1-treated cells. In conclusion, these results indicate that HPCs appear to differentiate into myofibroblasts and exhibit a profibrotic effect in progressive cirrhosis via activation of the non-canonical Wnt pathway. Blocking the non-canonical Wnt pathway can inhibit the differentiation of HPCs into myofibroblasts, suggesting that blocking this pathway and changing the fate of differentiated HPCs may be a potential treatment for cirrhosis.
Collapse
Affiliation(s)
- Jiamei Chen
- Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Institute of Liver Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiao Zhang
- Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Institute of Liver Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ying Xu
- Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Institute of Liver Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xuewei Li
- Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Institute of Liver Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shuang Ren
- Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Institute of Liver Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yaning Zhou
- Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Institute of Liver Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuyou Duan
- Department of Internal Medicine, Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, United States of America
| | - Mark Zern
- Department of Internal Medicine, Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, United States of America
| | - Hua Zhang
- Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Institute of Liver Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Gaofeng Chen
- Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Institute of Liver Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chenghai Liu
- Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Institute of Liver Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yongping Mu
- Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Institute of Liver Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ping Liu
- Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Institute of Liver Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
55
|
Mercer LK, Lunt M, Low ALS, Dixon WG, Watson KD, Symmons DPM, Hyrich KL. Risk of solid cancer in patients exposed to anti-tumour necrosis factor therapy: results from the British Society for Rheumatology Biologics Register for Rheumatoid Arthritis. Ann Rheum Dis 2015; 74:1087-93. [PMID: 24685910 PMCID: PMC4431340 DOI: 10.1136/annrheumdis-2013-204851] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 02/24/2014] [Accepted: 03/01/2014] [Indexed: 12/21/2022]
Abstract
BACKGROUND Patients with rheumatoid arthritis (RA) have an increased risk of certain solid cancers, in particular lung cancer, compared to the general population. Treatment with tumour necrosis factor (TNF) inhibitors (TNFi) may further enhance this risk. OBJECTIVES To compare the risk of solid cancer in patients with RA treated with TNFi to that in patients treated with non-biologic (synthetic) disease modifying antirheumatic drugs (sDMARDs). METHODS Patients with a physician diagnosis of RA enrolled in the British Society for Rheumatology Biologics Register, a national prospective cohort study established in 2001 to monitor the long-term safety of TNFi, were followed via record linkage with the national cancer registries until first solid cancer, death, for 5 years, or until 2011. Rates of solid cancers in 11 767 patients without prior cancer who received TNFi were compared to those in 3249 patients without prior cancer treated with sDMARDs. RESULTS 427 solid cancers were reported in 52 549 patient-years follow-up for the TNFi group (81 (95% CI 74 to 89) per 10 000 patient-years) and 136 cancers were reported in 11 672 patient-years in the sDMARD cohort (117 (95% CI 98 to 138) per 10 000 patient-years). After adjusting for differences in baseline characteristics there was no difference in risk of solid cancer for TNFi compared to sDMARD treated patients: HR 0.83 (95% CI 0.64 to 1.07). There was no difference in the relative risk of cancer for any of the individual TNFi drugs. CONCLUSIONS The addition of TNFi to sDMARD does not alter the risk of cancer in RA patients selected for TNFi in the UK.
Collapse
Affiliation(s)
- Louise K Mercer
- Arthritis Research UK Centre for Epidemiology, Centre for Musculoskeletal Research, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Mark Lunt
- Arthritis Research UK Centre for Epidemiology, Centre for Musculoskeletal Research, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Audrey L S Low
- Arthritis Research UK Centre for Epidemiology, Centre for Musculoskeletal Research, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - William G Dixon
- Arthritis Research UK Centre for Epidemiology, Centre for Musculoskeletal Research, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Kath D Watson
- Arthritis Research UK Centre for Epidemiology, Centre for Musculoskeletal Research, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Deborah P M Symmons
- Arthritis Research UK Centre for Epidemiology, Centre for Musculoskeletal Research, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Kimme L Hyrich
- Arthritis Research UK Centre for Epidemiology, Centre for Musculoskeletal Research, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| |
Collapse
|
56
|
Abstract
The link between chronic inflammation and cancer has long been suspected, due to the pioneering work of Rudolf Virchow over 150 years ago. Yet the causal relationship between inflammation and cancer was only deciphered in the past decade or so, using animal models of various cancers. Up to 20% of all human cancers result from chronic inflammation and persistent infections. Proinflammatory cytokines and tumor-infiltrating myeloid and immune cells play critical roles in almost every developmental stages of inflammation-induced cancers, from initiation, promotion, and progression to malignant metastasis. However, even in cancers with no preceding inflammation, inflammatory cells infiltrate tumor stroma and contribute to cancer development. Such "tumor-elicited inflammation" further emphasizes the importance of inflammation in different types of cancers, including that of the colon. In this review, we summarize our current knowledge of the function and induction mechanisms of inflammatory cytokines during colorectal cancer development, and hope to provide insight into the development of novel anticancer therapies by modulating tumor-elicited inflammation.
Collapse
Affiliation(s)
- Kepeng Wang
- Departments of Pharmacology and Pathology, Laboratory of Gene Regulation and Signal Transduction, School of Medicine, University of California, San Diego, California, USA
| | - Michael Karin
- Departments of Pharmacology and Pathology, Laboratory of Gene Regulation and Signal Transduction, School of Medicine, University of California, San Diego, California, USA.
| |
Collapse
|
57
|
Lebrec H, Ponce R, Preston BD, Iles J, Born TL, Hooper M. Tumor necrosis factor, tumor necrosis factor inhibition, and cancer risk. Curr Med Res Opin 2015; 31:557-74. [PMID: 25651481 DOI: 10.1185/03007995.2015.1011778] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
OBJECTIVE Tumor necrosis factor (TNF) is a highly pleiotropic cytokine with multiple activities other than its originally discovered role of tumor necrosis in rodents. TNF is now understood to play a contextual role in driving either tumor elimination or promotion. Using both animal and human data, this review examines the role of TNF in cancer development and the effect of TNF and TNF inhibitors (TNFis) on malignancy risk. RESEARCH DESIGN A literature review was performed using relevant search terms for TNF and malignancy. RESULTS Although administration of TNF can cause tumor regression in specific rodent tumor models, human expression polymorphisms suggest that TNF can be a tumor-promoting cytokine, whereas blocking the TNF pathway in a variety of tumor models inhibits tumor growth. In addition to direct effects of TNF on tumors, TNF can variously affect immunity and the tumor microenvironment. Whereas TNF can promote immune surveillance designed to eliminate tumors, it can also drive chronic inflammation, autoimmunity, angiogenesis, and other processes that promote tumor initiation, growth, and spread. Key players in TNF signaling that shape this response include NF-κB and JNK, and malignant-inflammatory cell interactions, each of which may have different responses to TNF signaling. Focusing on rheumatoid arthritis (RA) patients, where clinical experience is most extensive, a review of the clinical literature shows no increased risk of overall malignancy or solid tumors such as breast and lung cancers with exposure to TNFis. Lymphoma rates are not increased with use of TNFis. Conflicting data exist regarding the risks of melanoma and nonmelanoma skin cancer. Data regarding the risk of recurrent malignancy are limited. CONCLUSIONS Overall, the available data indicate that elevated TNF is a risk factor for cancer, whereas its inhibition in RA patients is not generally associated with an increased cancer risk. In particular, TNF inhibition is not associated with cancers linked to immune suppression. A better understanding of the tumor microenvironment, molecular events underlying specific tumors, and epidemiologic studies of malignancies within specific disease indications should enable more focused pharmacovigilance studies and a better understanding of the potential risks of TNFis.
Collapse
|
58
|
Liu X, Zhang J, Li J, Volk A, Breslin P, Zhang J, Zhang Z. The synergistic repressive effect of NF-κB and JNK inhibitor on the clonogenic capacity of Jurkat leukemia cells. PLoS One 2014; 9:e115490. [PMID: 25526629 PMCID: PMC4272284 DOI: 10.1371/journal.pone.0115490] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 11/24/2014] [Indexed: 12/22/2022] Open
Abstract
Deregulation of Nuclear Transcription Factor-κB (NF-κB) and Jun N-terminal kinase (JNK) signaling is commonly detected in leukemia, suggesting an important role for these two signaling pathways in the pathogenesis of leukemia. In this study, using Jurkat cells, an acute T-lymphoblastic leukemia (T-ALL) cell line, we evaluated the effects of an NF-κB inhibitor and a JNK inhibitor individually and in combination on the proliferation, survival and clonogenic capacity of leukemic cells. We found that leukemic stem/progenitor cells (LSPCs) were more sensitive to NF-κB inhibitor treatment than were healthy hematopoietic stem/progenitor cells (HSPCs), as shown by a reduction in the clonogenic capacity of the former. Inactivation of NF-κB leads to the activation of JNK signaling in both leukemic cells and healthy HSPCs. Interestingly, JNK inhibitor treatment enhanced the repressive effects of NF-κB inhibitor on LSPCs but prevented such repression in HSPCs. Our data suggest that JNK signaling stimulates proliferation/survival in LSPCs but is a death signal in HSPCs. The combination of NF-κB inhibitor and JNK inhibitor might provide a better treatment for T-ALL leukemia by synergistically killing LSPCs while simultaneously preventing the death of normal HPCs.
Collapse
Affiliation(s)
- Xinli Liu
- Department of Biology, College of Life and Environment Science, Shanghai Normal University, Shanghai, People's Republic of China
| | - Jun Zhang
- Department of Biology, College of Life and Environment Science, Shanghai Normal University, Shanghai, People's Republic of China
| | - Jing Li
- Department of Biology, College of Life and Environment Science, Shanghai Normal University, Shanghai, People's Republic of China
| | - Andrew Volk
- Oncology Institute, Cardinal Bernardin Cancer Center and Departments of Pathology and Molecular/Cellular Physiology, Loyola University Chicago, Maywood, IL 60153, United States of America
| | - Peter Breslin
- Oncology Institute, Cardinal Bernardin Cancer Center and Departments of Pathology and Molecular/Cellular Physiology, Loyola University Chicago, Maywood, IL 60153, United States of America
| | - Jiwang Zhang
- Oncology Institute, Cardinal Bernardin Cancer Center and Departments of Pathology and Molecular/Cellular Physiology, Loyola University Chicago, Maywood, IL 60153, United States of America
| | - Zhou Zhang
- College of Life Science and Biopharmacy, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
- * E-mail:
| |
Collapse
|
59
|
Liu D, Yovchev MI, Zhang J, Alfieri AA, Tchaikovskaya T, Laconi E, Dabeva MD. Identification and characterization of mesenchymal-epithelial progenitor-like cells in normal and injured rat liver. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 185:110-28. [PMID: 25447047 DOI: 10.1016/j.ajpath.2014.08.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 08/22/2014] [Accepted: 08/27/2014] [Indexed: 01/07/2023]
Abstract
In normal rat liver, thymocyte antigen 1 (Thy1) is expressed in fibroblasts/myofibroblasts and in some blood progenitor cells. Thy1-expressing cells also accumulate in the liver during impaired liver regeneration. The origin and nature of these cells are not well understood. By using RT-PCR analysis and immunofluorescence microscopy, we describe the presence of rare Thy1(+) cells in the liver lobule of normal animals, occasionally forming small collections of up to 20 cells. These cells constitute a small portion (1.7% to 1.8%) of nonparenchymal cells and reveal a mixed mesenchymal-epithelial phenotype, expressing E-cadherin, cytokeratin 18, and desmin. The most potent mitogens for mesenchymal-epithelial Thy1(+) cells in vitro are the inflammatory cytokines interferon γ, IL-1, and platelet-derived growth factor-BB, which are not produced by Thy1(+) cells. Thy1(+) cells express all typical mesenchymal stem cell and hepatic progenitor cell markers and produce growth factor and cytokine mRNA (Hgf, Il6, Tgfa, and Tweak) for proteins that maintain oval cell growth and differentiation. Under appropriate conditions, mesenchymal-epithelial cells differentiate in vitro into hepatocyte-like cells. In this study, we show that the adult rat liver harbors a small pool of endogenous mesenchymal-epithelial cells not recognized previously. In the quiescent state, these cells express both mesenchymal and epithelial cell markers. They behave like hepatic stem cells/progenitors with dual phenotype, exhibiting high plasticity and long-lasting proliferative activity.
Collapse
Affiliation(s)
- Daqing Liu
- Department of Medicine, Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, New York
| | - Mladen I Yovchev
- Department of Medicine, Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, New York
| | - Jinghang Zhang
- Flow Cytometry Core Facility, Albert Einstein College of Medicine, Bronx, New York
| | - Alan A Alfieri
- Department of Radiation Oncology, Albert Einstein College of Medicine, Bronx, New York
| | - Tatyana Tchaikovskaya
- Department of Medicine, Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, New York
| | - Ezio Laconi
- Section of Experimental Pathology, Department of Sciences and Biomedical Technology, University of Cagliari, Cagliari, Italy
| | - Mariana D Dabeva
- Department of Medicine, Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, New York.
| |
Collapse
|
60
|
O'Rourke RW. Obesity and cancer: at the crossroads of cellular metabolism and proliferation. Surg Obes Relat Dis 2014; 10:1208-19. [PMID: 25264328 PMCID: PMC4267907 DOI: 10.1016/j.soard.2014.08.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 08/19/2014] [Accepted: 08/21/2014] [Indexed: 12/14/2022]
Abstract
Obesity is associated with an increased risk of cancer. The mechanisms underlying this association include but are not limited to increased systemic inflammation, an anabolic hormonal milieu, and adipocyte-cancer crosstalk, aberrant stimuli that conspire to promote neoplastic transformation. Cellular proliferation is uncoupled from nutrient availability in malignant cells, promoting tumor progression. Elucidation of the mechanisms underlying the obesity-cancer connection will lead to the development of novel metabolism-based agents for cancer prevention and treatment.
Collapse
Affiliation(s)
- Robert W O'Rourke
- Department of Surgery, University of Michigan and Ann Arbor VA Hospital, Ann Arbor, Michigan.
| |
Collapse
|
61
|
TIMP3 controls cell fate to confer hepatocellular carcinoma resistance. Oncogene 2014; 34:4098-108. [PMID: 25347747 DOI: 10.1038/onc.2014.339] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 08/29/2014] [Accepted: 09/04/2014] [Indexed: 02/07/2023]
Abstract
Inflammation enables human cancers and is a critical promoter of hepatocellular carcinoma (HCC). TIMP3 (Tissue inhibitor of metalloproteinase 3), a natural metalloproteinase inhibitor, controls cytokine and growth factor bioavailability to keep inflammation in check and regulate cell survival in the liver. TIMP3 is also found silenced in human cancers. We therefore tested whether Timp3 affects HCC predisposition. Remarkably, genetic loss of Timp3 protected from carcinogen-induced HCC through the immediate engagement of several tumor suppressor pathways, while tumor necrosis factor (TNF) signaling was dispensable for this protection. All wild-type mice developed HCC by 12 months, whereas HCC incidence was reduced to 33% at 12 months and 57% at 15 months in Timp3 null mice. Upon acute carcinogen treatment the deficient livers exhibited greater cytokine expression, but lower cell death and higher hepatocyte senescence. We found that precocious activation of p53, p38 and Notch preceded senescence and hepatic cell differentiation, and these events were conserved throughout tumorigenesis. Timp3-deficient mouse embryo fibroblasts also responded to carcinogen by favoring senescence over apoptosis. We conclude that Timp3 status determines p53, p38 and Notch coactivation to instruct hepatic cell fate and transformation and uncover mechanisms that are protective even within a pro-inflammatory microenvironment.
Collapse
|
62
|
De Minicis S, Marzioni M, Benedetti A, Svegliati-Baroni G. New insights in hepatocellular carcinoma: from bench to bedside. ANNALS OF TRANSLATIONAL MEDICINE 2014; 1:15. [PMID: 25332959 DOI: 10.3978/j.issn.2305-5839.2013.01.06] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Accepted: 01/31/2013] [Indexed: 01/10/2023]
Abstract
Hepatocarcinogenesis is a multistep process involving different genetic alterations that ultimately lead to malignant transformation of the hepatocyte. The liver is one of the main targets for different metastatic foci, but it represents an important and frequent locus of degeneration in the course of chronic disease. In fact, Hepatocellular carcinoma (HCC) represents the outcome of the natural history of chronic liver diseases, from the condition of fibrosis, to cirrhosis and finally to cancer. HCC is the sixth most common cancer in the world, some 630,000 new cases being diagnosed each year. Furthermore, about the 80% of people with HCC, have seen their clinical history developing from fibrosis, to cirrhosis and finally to cancer. The three main causes of HCC development are represented by HBV, HCV infection and alcoholism. Moreover, metabolic disease [starting from Non Alcoholic Fatty Liver Disease (NAFLD), Non Alcoholic Steatohepatitis (NASH)] and, with reduced frequency, some autoimmune disease may lead to HCC development. An additional rare cause of carcinogenetic degeneration of the liver, especially developed in African and Asian Countries, is represented by aflatoxin B1. The mechanisms by which these etiologic factors may induce HCC development involve a wide range of pathway and molecules, currently under investigation. In summary, the hepatocarcionogenesis results from a multifactorial process leading to the common condition of genetic changes in mature hepatocytes mainly characterized by uncontrolled proliferation and cell death. Advances in understanding the mechanism of action are fundamental for the development of new potential therapies and results primarily from the association of the research activities coming from basic and clinical science. This review article analyzes the current models used in basic research to investigate HCC activity, and the advances obtained from a basic and clinical point of view.
Collapse
Affiliation(s)
- Samuele De Minicis
- Department of Gastroenterology, Università Politecnica delle Marche, Ancona, Italy
| | - Marco Marzioni
- Department of Gastroenterology, Università Politecnica delle Marche, Ancona, Italy
| | - Antonio Benedetti
- Department of Gastroenterology, Università Politecnica delle Marche, Ancona, Italy
| | | |
Collapse
|
63
|
Neonatal streptozotocin treatment causes type 1 diabetes and subsequent hepatocellular carcinoma in DIAR mice fed a normal diet. Hepatol Int 2014. [DOI: 10.1007/s12072-014-9541-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
64
|
Sharma M, Mohapatra J, Argade A, Deshpande SS, Shah GB, Chatterjee A, Jain MR. Chemopreventive effect of a novel, selective TACE inhibitor on DMBA- and TPA-induced skin carcinogenesis. Immunopharmacol Immunotoxicol 2014; 36:282-9. [PMID: 24946851 DOI: 10.3109/08923973.2014.931421] [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] [Indexed: 12/14/2022]
Abstract
UNLABELLED Abstract Context: Tumor necrosis factor (TNF)-α, a potent proinflammatory cytokine, plays a major role in the pathogenesis of cancer. TNF-α converting enzyme (TACE) mediates processing and release of biologically active TNF-α. OBJECTIVE We aimed to investigate the effect of a novel, selective TACE inhibitor (compound 11p) on skin inflammation and associated tumorigenesis in mice. METHODS Skin edema was induced in mice by dermal application 12-O-tetradecanoylphorbol-13-acetate (TPA) solution in acetone on to the ear and the effect of post-treatment of compound 11p (topical application) was evaluated. Edema and inflammation was assessed by measuring ear thickness, weight of skin punch and cytokine levels. Skin cancer in mice was initiated by single topical application of 7,12-dimethylbenz[a]anthracene (DMBA) and promoted by repeated TPA application for 20 weeks. The effect of compound 11p on papilloma incidence and multiplicity was evaluated. RESULTS Treatment with compound 11p strongly suppressed TPA-induced elevation in skin thickness and weight. A dose-dependent suppression in TPA-mediated TNF-α, IL-6, IFN-γ, IL-17 and PGE2 levels which was associated with a decrease in infiltration of inflammatory cells was also observed with the treatment. Moreover, compound 11p treatment delayed the onset, markedly reduced the papilloma incidence and multiplicity persuaded by DMBA and TPA. DISCUSSION AND CONCLUSION These findings suggest that selective blockade of TACE suppresses TPA-induced epidermal hyperplasia, inflammatory cell infiltration and cytokine level. Inhibition of inflammatory events related to tumor growth might have led to the anti-tumor effect in mouse skin cancer model induced by DMBA and TPA.
Collapse
|
65
|
Volk A, Li J, Xin J, You D, Zhang J, Liu X, Xiao Y, Breslin P, Li Z, Wei W, Schmidt R, Li X, Zhang Z, Kuo PC, Nand S, Zhang J, Chen J, Zhang J. Co-inhibition of NF-κB and JNK is synergistic in TNF-expressing human AML. ACTA ACUST UNITED AC 2014; 211:1093-108. [PMID: 24842373 PMCID: PMC4042653 DOI: 10.1084/jem.20130990] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Leukemic stem cells (LSCs) isolated from acute myeloid leukemia (AML) patients are more sensitive to nuclear factor κB (NF-κB) inhibition-induced cell death when compared with hematopoietic stem and progenitor cells (HSPCs) in in vitro culture. However, inadequate anti-leukemic activity of NF-κB inhibition in vivo suggests the presence of additional survival/proliferative signals that can compensate for NF-κB inhibition. AML subtypes M3, M4, and M5 cells produce endogenous tumor necrosis factor α (TNF). Although stimulating HSPC with TNF promotes necroptosis and apoptosis, similar treatment with AML cells (leukemic cells, LCs) results in an increase in survival and proliferation. We determined that TNF stimulation drives the JNK-AP1 pathway in a manner parallel to NF-κB, leading to the up-regulation of anti-apoptotic genes in LC. We found that we can significantly sensitize LC to NF-κB inhibitor treatment by blocking the TNF-JNK-AP1 signaling pathway. Our data suggest that co-inhibition of both TNF-JNK-AP1 and NF-κB signals may provide a more comprehensive treatment paradigm for AML patients with TNF-expressing LC.
Collapse
Affiliation(s)
- Andrew Volk
- Molecular Biology Program, Department of Biology, Loyola University Chicago, Chicago, IL 60660
| | - Jing Li
- Department of Biology, College of Life and Environment Science, Shanghai Normal University, Shanghai 200234, People's Republic of China
| | - Junping Xin
- Oncology Institute, Cardinal Bernardin Cancer Center, Department of Pathology; Department of Molecular and Cellular Physiology; and Department of Medicine, Loyola University Medical Center, Maywood, IL 60153
| | - Dewen You
- Oncology Institute, Cardinal Bernardin Cancer Center, Department of Pathology; Department of Molecular and Cellular Physiology; and Department of Medicine, Loyola University Medical Center, Maywood, IL 60153
| | - Jun Zhang
- Department of Biology, College of Life and Environment Science, Shanghai Normal University, Shanghai 200234, People's Republic of China
| | - Xinli Liu
- Department of Biology, College of Life and Environment Science, Shanghai Normal University, Shanghai 200234, People's Republic of China
| | - Yechen Xiao
- Oncology Institute, Cardinal Bernardin Cancer Center, Department of Pathology; Department of Molecular and Cellular Physiology; and Department of Medicine, Loyola University Medical Center, Maywood, IL 60153
| | - Peter Breslin
- Molecular Biology Program, Department of Biology, Loyola University Chicago, Chicago, IL 60660 Oncology Institute, Cardinal Bernardin Cancer Center, Department of Pathology; Department of Molecular and Cellular Physiology; and Department of Medicine, Loyola University Medical Center, Maywood, IL 60153 Oncology Institute, Cardinal Bernardin Cancer Center, Department of Pathology; Department of Molecular and Cellular Physiology; and Department of Medicine, Loyola University Medical Center, Maywood, IL 60153
| | - Zejuan Li
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Wei Wei
- Oncology Institute, Cardinal Bernardin Cancer Center, Department of Pathology; Department of Molecular and Cellular Physiology; and Department of Medicine, Loyola University Medical Center, Maywood, IL 60153
| | - Rachel Schmidt
- Oncology Institute, Cardinal Bernardin Cancer Center, Department of Pathology; Department of Molecular and Cellular Physiology; and Department of Medicine, Loyola University Medical Center, Maywood, IL 60153
| | - Xingyu Li
- Department of Biology, College of Life and Environment Science, Shanghai Normal University, Shanghai 200234, People's Republic of China
| | - Zhou Zhang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Paul C Kuo
- Oncology Institute, Cardinal Bernardin Cancer Center, Department of Pathology; Department of Molecular and Cellular Physiology; and Department of Medicine, Loyola University Medical Center, Maywood, IL 60153
| | - Sucha Nand
- Oncology Institute, Cardinal Bernardin Cancer Center, Department of Pathology; Department of Molecular and Cellular Physiology; and Department of Medicine, Loyola University Medical Center, Maywood, IL 60153
| | - Jianke Zhang
- Thomas Jefferson University, Jefferson Medical College, Department of Microbiology and Immunology, Philadelphia, PA 19107
| | - Jianjun Chen
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Jiwang Zhang
- Molecular Biology Program, Department of Biology, Loyola University Chicago, Chicago, IL 60660 Oncology Institute, Cardinal Bernardin Cancer Center, Department of Pathology; Department of Molecular and Cellular Physiology; and Department of Medicine, Loyola University Medical Center, Maywood, IL 60153
| |
Collapse
|
66
|
Stem/Progenitor Cells in Liver Development, Homeostasis, Regeneration, and Reprogramming. Cell Stem Cell 2014; 14:561-74. [DOI: 10.1016/j.stem.2014.04.010] [Citation(s) in RCA: 384] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
67
|
Itoh T, Miyajima A. Liver regeneration by stem/progenitor cells. Hepatology 2014; 59:1617-26. [PMID: 24115180 DOI: 10.1002/hep.26753] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 09/11/2013] [Indexed: 12/12/2022]
Abstract
UNLABELLED The liver is renowned for its strong, robust regenerative capacity, employing different modes of regeneration according to type and extent of injury. The process of compensatory hypertrophy of the liver upon partial hepatectomy has been standing as a classical model for studying organ regeneration in mammals and a subject of exhaustive analyses. Meanwhile, in view of the physiological relevance for many of the human liver pathologies induced upon toxic insults or hepatitis, other injury models have recently drawn increasing attention. In those damaged livers where hepatocyte proliferation is compromised, adult liver stem/progenitor cells (LPCs) are activated and differentiate to hepatocytes and cholangiocytes, leading to functional recovery of the organ. Here, we summarize and discuss recent findings on the mechanisms underlying the regeneration process of the liver. Whereas the primary focus of this article is on those related to LPC-mediated regeneration, we also introduce topics on compensatory hypertrophy, where application of new technologies and molecular genetics approaches in mice has gained a paradigm shift. Identification of various markers for LPC populations has expedited their characterization and enabled us to examine their differentiation potential in vivo using genetic lineage-tracing approaches. Comprehensive studies regarding intercellular signaling pathways and their modes of action have succeeded in elucidating novel frameworks for the LPC-niche interaction functioning in the regenerating liver. CONCLUSION Advancing our understanding of the cellular and molecular mechanisms for liver regeneration should provide a basis for developing therapeutic strategies to treat patients with liver disease.
Collapse
Affiliation(s)
- Tohru Itoh
- Laboratory of Cell Growth and Differentiation, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo, Japan
| | | |
Collapse
|
68
|
Wang HY, Yang SL, Liang HF, Li CH. HBx protein promotes oval cell proliferation by up-regulation of cyclin D1 via activation of the MEK/ERK and PI3K/Akt pathways. Int J Mol Sci 2014; 15:3507-18. [PMID: 24577313 PMCID: PMC3975350 DOI: 10.3390/ijms15033507] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 02/18/2014] [Accepted: 02/18/2014] [Indexed: 12/13/2022] Open
Abstract
Growing evidence has shown that hepatic oval cells, also named liver progenitor cells, play an important role in the process of liver regeneration in various liver diseases. Oval cell proliferation has been reported in hepatitis B virus (HBV)-associated hepatocellular carcinoma (HCC) and chronic liver disease. Studies have found expression of HBV surface and core antigens in oval cells in the livers of patients with HCC, suggesting that HBV infection of oval cells could be a mechanism of human hepatocarcinogenesis. In addition, there is evidence of multiplication of HBV in oval cell culture. However, little research has been performed to explore the role of HBV-encoded proteins in the proliferation of hepatic oval cells. Previously, we successfully transfected the HBV x (HBx) gene, one of the four genes in the HBV genome, into a rat LE/6 oval cell line. In this study, we tested whether or not the transfected HBx gene could affect oval cell proliferation in vitro. Our results show that overexpression of HBx promotes the proliferation of oval cells and increases cyclin D1 expression, assessed at both the mRNA and protein levels. We also found that HBx activated the PI-3K/Akt and MEK/ERK1/2 pathways in HBx-transfected oval cells. Furthermore, the HBx-induced increases in cyclin D1 expression and oval cell proliferation were completely abolished by treatment with either MEK inhibitor PD184352 or PI-3K inhibitor LY294002. These results demonstrated that HBx has the ability to promote oval cell proliferation in vitro, and its stimulatory effects on cell proliferation and expression of cyclin D1 depend on the activation of the MEK/ERK and PI3K/Akt signaling pathways in cultured oval cells.
Collapse
Affiliation(s)
- Heng-Yi Wang
- Department of Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China.
| | - Sheng-Li Yang
- Department of General Surgery, Liyuan Hospital, Tongji Medical College, Huazhong Science and Technology University, Wuhan 430077, China.
| | - Hui-Fang Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong Science and Technology University, Wuhan 430030, China.
| | - Chang-Hai Li
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong Science and Technology University, Wuhan 430030, China.
| |
Collapse
|
69
|
Schotanus BA, Penning LC, Spee B. Potential of regenerative medicine techniques in canine hepatology. Vet Q 2014; 33:207-16. [PMID: 24422896 DOI: 10.1080/01652176.2013.875240] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Liver cell turnover is very slow, especially compared to intestines and stomach epithelium and hair cells. Since the liver is the main detoxifying organ in the body, it does not come as a surprise that the liver has an unmatched regenerative capacity. After 70% partial hepatectomy, the liver size returns to normal in about two weeks due to replication of differentiated hepatocytes and cholangiocytes. Despite this, liver diseases are regularly encountered in the veterinary clinic. Dogs primarily present with parenchymal pathologies such as hepatitis. The estimated frequency of canine hepatitis depends on the investigated population and accounts for 1%-2% of our university clinic referral population, and up to 12% in a general population. In chronic and severe acute liver disease, the regenerative and replicative capacity of the hepatocytes and/or cholangiocytes falls short and the liver is not restored. In this situation, proliferation of hepatic stem cells or hepatic progenitor cells (HPCs), on histology called the ductular reaction, comes into play to replace the damaged hepatocytes or cholangiocytes. For unknown reasons the ductular reaction is often too little and too late, or differentiation into fully differentiated hepatocytes or cholangiocytes is hampered. In this way, HPCs fail to fully regenerate the liver. The presence and potential of HPCs does, however, provide great prospectives for their use in regenerative strategies. This review highlights the regulation of, and the interaction between, HPCs and other liver cell types and discusses potential regenerative medicine-oriented strategies in canine hepatitis, making use of (liver) stem cells.
Collapse
Affiliation(s)
- Baukje A Schotanus
- a Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine , Utrecht University , Utrecht , The Netherlands
| | | | | |
Collapse
|
70
|
Chen CP, Haas-Kogan D. Neoplasms of the hepatobiliary system: clinical presentation, molecular pathways and diagnostics. Expert Rev Mol Diagn 2014; 10:883-95. [DOI: 10.1586/erm.10.76] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
71
|
Chaudhary K, Liedtke C, Wertenbruch S, Trautwein C, Streetz KL. Caspase 8 differentially controls hepatocytes and non-parenchymal liver cells during chronic cholestatic liver injury in mice. J Hepatol 2013; 59:1292-8. [PMID: 23928400 DOI: 10.1016/j.jhep.2013.07.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2012] [Revised: 07/14/2013] [Accepted: 07/16/2013] [Indexed: 01/08/2023]
Abstract
BACKGROUND & AIMS Receptor mediated cell death through the activation of caspases has been identified as an important mechanism to control life and death in various tissues and is thus crucial for the maintenance of liver tissue homeostasis. Here we investigated how caspase 8 (Casp8) differentially regulates immune-mediated liver injury and regeneration in distinct liver cell types during chronic liver injury. METHODS Conditional knockout mice with hepatocellular (Casp8(Δhepa)) and ubiquitous deletion of Casp8 (Casp8(ΔMx)) were used in models of cholestatic hepatitis [(DDC (3,5-diethoxycarbonyl-1,4-dihydrocollidine) treatment, bile duct ligation (BDL) and choline deficient diet with ethionine supplementation (CDE)]. RESULTS Mice with a hepatocellular deletion of Casp8 (Casp8(Δhepa)) were protected after DDC-treatment. Animals with a ubiquitous conditional Casp8 knockout (Casp8(ΔMx)) displayed a significantly enhanced liver injury in various models of cholestatic liver injury. This was associated with higher transaminases, bilirubin levels and finally more liver fibrosis. However, caspase 3 (Casp3) activity was reduced in both knockout strains, suggesting additionally mechanisms contributing to the phenotype. Casp8(ΔMx) mice displayed a stronger infiltration of mononuclear immune cells and more proliferation of liver-parenchymal cells in periportal areas. Further analysis confirmed that these infiltrating immune cells are resistant against extrinsic apoptosis. Bone-marrow-transplantation (BMT) experiments demonstrated that Casp8-deficient bone marrow derived cells are responsible for increased liver injury in DDC fed mice. CONCLUSIONS Our results demonstrate that cell-type specific differences in apoptosis resistance mediated by Casp8 deletion are of significant relevance for the outcome of chronic liver injury.
Collapse
Affiliation(s)
- Kunal Chaudhary
- Department of Medicine III, University Hospital Aachen, RWTH Aachen University, Pauwelsstrasse 30, 52074 Aachen, Germany
| | | | | | | | | |
Collapse
|
72
|
Zhang WJ, Guo Y. Mechanisms of liver repair following injury. Shijie Huaren Xiaohua Zazhi 2013; 21:3369-3375. [DOI: 10.11569/wcjd.v21.i31.3369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Liver injury caused by a variety of physical or chemical factors is a common disease, and severe or persistent liver injury can ultimately lead to acute liver failure. Its treatment is still a formidable challenge to clinicians. Elucidation of mechanisms underlying liver repair following injury is the cornerstone of treatment of hepatic diseases. Despite many research efforts over the past decades, the mechanisms behind liver repair following injury are still not clear. Recent studies have demonstrated that oval cells and bone marrow stem cells are involved in this complex process. A variety of cells and factors may play a role in different stages of this process. In this paper, we will review mechanisms of liver repair following injury.
Collapse
|
73
|
Cubero FJ, Singh A, Borkham-Kamphorst E, Nevzorova YA, Al Masaoudi M, Haas U, Boekschoten MV, Gassler N, Weiskirchen R, Muller M, Liedtke C, Trautwein C. TNFR1 determines progression of chronic liver injury in the IKKγ/Nemo genetic model. Cell Death Differ 2013; 20:1580-1592. [PMID: 23933814 PMCID: PMC3792433 DOI: 10.1038/cdd.2013.112] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Revised: 06/18/2013] [Accepted: 07/12/2013] [Indexed: 12/26/2022] Open
Abstract
Death receptor-mediated hepatocyte apoptosis is implicated in a wide range of liver diseases including viral and alcoholic hepatitis, ischemia/reperfusion injury, fulminant hepatic failure, cholestatic liver injury, as well as cancer. Deletion of NF-κB essential modulator in hepatocytes (IKKγ/Nemo) causes spontaneous progression of TNF-mediated chronic hepatitis to hepatocellular carcinoma (HCC). Thus, we analyzed the role of death receptors including TNFR1 and TRAIL in the regulation of cell death and the progression of liver injury in IKKγ/Nemo-deleted livers. We crossed hepatocyte-specific IKKγ/Nemo knockout mice (Nemo(Δhepa)) with constitutive TNFR1(-/-) and TRAIL(-/-) mice. Deletion of TNFR1, but not TRAIL, decreased apoptotic cell death, compensatory proliferation, liver fibrogenesis, infiltration of immune cells as well as pro-inflammatory cytokines, and indicators of tumor growth during the progression of chronic liver injury. These events were associated with diminished JNK activation. In contrast, deletion of TNFR1 in bone-marrow-derived cells promoted chronic liver injury. Our data demonstrate that TNF- and not TRAIL signaling determines the progression of IKKγ/Nemo-dependent chronic hepatitis. Additionally, we show that TNFR1 in hepatocytes and immune cells have different roles in chronic liver injury-a finding that has direct implications for treating chronic liver disease.
Collapse
Affiliation(s)
- F J Cubero
- Department of Internal Medicine III, University Hospital, RWTH Aachen, Aachen, Germany
| | - A Singh
- Department of Internal Medicine III, University Hospital, RWTH Aachen, Aachen, Germany
| | - E Borkham-Kamphorst
- Institute of Clinical Chemistry and Pathobiochemistry, University Hospital, RWTH Aachen, Aachen, Germany
| | - Y A Nevzorova
- Department of Internal Medicine III, University Hospital, RWTH Aachen, Aachen, Germany
| | - M Al Masaoudi
- Department of Internal Medicine III, University Hospital, RWTH Aachen, Aachen, Germany
| | - U Haas
- Institute of Clinical Chemistry and Pathobiochemistry, University Hospital, RWTH Aachen, Aachen, Germany
| | - M V Boekschoten
- Division of Human Nutrition, Metabolism and Genomics, Wageningen University, Wageningen, The Netherlands
| | - N Gassler
- Institute of Pathology, University Hospital, RWTH Aachen, Germany
| | - R Weiskirchen
- Institute of Clinical Chemistry and Pathobiochemistry, University Hospital, RWTH Aachen, Aachen, Germany
| | - M Muller
- Division of Human Nutrition, Metabolism and Genomics, Wageningen University, Wageningen, The Netherlands
| | - C Liedtke
- Department of Internal Medicine III, University Hospital, RWTH Aachen, Aachen, Germany
| | - C Trautwein
- Department of Internal Medicine III, University Hospital, RWTH Aachen, Aachen, Germany
| |
Collapse
|
74
|
Rodríguez-Berriguete G, Sánchez-Espiridión B, Cansino JR, Olmedilla G, Martínez-Onsurbe P, Sánchez-Chapado M, Paniagua R, Fraile B, Royuela M. Clinical significance of both tumor and stromal expression of components of the IL-1 and TNF-α signaling pathways in prostate cancer. Cytokine 2013; 64:555-63. [DOI: 10.1016/j.cyto.2013.09.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 07/05/2013] [Accepted: 09/01/2013] [Indexed: 10/26/2022]
|
75
|
Kou X, Jing Y, Deng W, Sun K, Han Z, Ye F, Yu G, Fan Q, Gao L, Zhao Q, Zhao X, Li R, Wei L, Wu M. Tumor necrosis factor-α attenuates starvation-induced apoptosis through upregulation of ferritin heavy chain in hepatocellular carcinoma cells. BMC Cancer 2013; 13:438. [PMID: 24066693 PMCID: PMC3849379 DOI: 10.1186/1471-2407-13-438] [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: 02/13/2013] [Accepted: 09/19/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Tumor microenviroment is characteristic of inflammation, ischemia and starvation of nutrient. TNF-α, which is an extraordinarily pleiotropic cytokine, could be an endogenous tumor promoter in some tumor types. The basic objective of this study was to investigate the effects of TNF-α on the cell viability and apoptosis of hepatocellular carcinoma cells under serum starvation, and to identify the molecular mechanisms involved. METHODS For this purpose, five different concentrations of TNF-α and two different serum settings (serum-cultured and serum-deprived) were used to investigate the effects of TNF-α on the cell viability and apoptosis of Hep3B and SMMC-7721 cells. RESULTS TNF-α (10 ng/ml) attenuated serum starvation-induced apoptosis of hepatocellular carcinoma cells, and autophagy conferred this process. BAY11-7082, a specific inhibitor of NF-κB, reversed the suppression of serum starvation-induced apoptosis by TNF-α. Moreover, TNF-α-induced NF-κB transactivation was suppressed by autophagy inhibitor 3-MA. In addition, TNF-α up-regulated Ferritin heavy chain (FHC) transiently by NF-κB activation and FHC levels were correlated with the TNF-α-induced protection against serum starvation-mediated apoptosis of hepatocellular carcinoma cells. Furthermore, FHC-mediated inhibition of apoptosis depended on suppressing ROS accumulation. CONCLUSIONS Our findings suggested that autophagy conferred the TNF-α protection against serum starvation-mediated apoptosis of hepatocellular carcinoma cells, the mechanism involved with the activation of the TNF-α/ NF-κB /FHC signaling pathway.
Collapse
Affiliation(s)
- Xingrui Kou
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, Shanghai 200438, P, R China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Alzahrani B, Iseli TJ, Hebbard LW. Non-viral causes of liver cancer: does obesity led inflammation play a role? Cancer Lett 2013; 345:223-9. [PMID: 24007864 DOI: 10.1016/j.canlet.2013.08.036] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 08/21/2013] [Accepted: 08/24/2013] [Indexed: 12/20/2022]
Abstract
Liver cancer is the fifth most common cancer worldwide and the third most common cause of cancer mortality. Hepatocellular carcinoma (HCC) accounts for around 90% of primary liver cancers. Chronic infection with hepatitis B and hepatitis C viruses are two of most common causes of liver cancer. However, there are non-viral factors that are associated with liver cancer development. Numerous population studies have revealed strong links between obesity and the development of liver cancer. Obesity can alter hepatic pathology, metabolism and promote inflammation, leading to nonalcoholic fatty liver disease (NAFLD) and the progression to the more severe form, non-alcoholic steatohepatitis (NASH). NASH is characterised by prominent steatosis and inflammation, and can lead to HCC. Here, we discuss the role of obesity in inflammation and the principal signalling mechanisms involved in HCC formation.
Collapse
Affiliation(s)
- Badr Alzahrani
- Storr Liver Unit, Westmead Millennium Institute and Westmead Hospital, University of Sydney, Darcy Road, Westmead, NSW 2145, Australia
| | - Tristan J Iseli
- Storr Liver Unit, Westmead Millennium Institute and Westmead Hospital, University of Sydney, Darcy Road, Westmead, NSW 2145, Australia
| | - Lionel W Hebbard
- Storr Liver Unit, Westmead Millennium Institute and Westmead Hospital, University of Sydney, Darcy Road, Westmead, NSW 2145, Australia.
| |
Collapse
|
77
|
Wu C, Wang X, Zhong M, Liu H, He Q, Yang X, Wen J, Feng D. Evaluation of potential reference genes for qRT-PCR studies in human hepatoma cell lines treated with TNF-α. Acta Biochim Biophys Sin (Shanghai) 2013; 45:780-6. [PMID: 23811755 DOI: 10.1093/abbs/gmt072] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
In this study, the expression of eight candidate reference genes, B2M, ACTB, GAPDH, HMBS, HPRT1, TBP, UBC, and YWHAZ, was examined to identify optimal reference genes by quantitative reverse transcription-polymerase chain reaction (qRT-PCR) analysis in two human hepatoma cell lines, BEL-7402 and SMMC-7721, treated with tumor necrosis factor-α (TNF-α) for different time periods. The expression stability of these genes was analyzed by three independent algorithms: geNorm, NormFinder, and BestKeeper. Results showed that TBP was the most stably expressed gene in BEL-7402 and SMMC-7721 cell lines under current experimental conditions, and that the optimal set of reference genes required for accurate normalization was TBP and HMBS, based on the pairwise variation value determined with geNorm. UBC and ACTB were ranked as the least stable genes by same algorithms. Our findings provide evidence that using TBP alone or in combination with HMBS as endogenous controls could be a reliable method for normalizing qRT-PCR data in human hepatoma cell lines treated with TNF-α.
Collapse
Affiliation(s)
- Chang Wu
- Department of Pathology, School of Basic Medical Sciences, Central South University, Changsha, China
| | | | | | | | | | | | | | | |
Collapse
|
78
|
Pusterla T, Nèmeth J, Stein I, Wiechert L, Knigin D, Marhenke S, Longerich T, Kumar V, Arnold B, Vogel A, Bierhaus A, Pikarsky E, Hess J, Angel P. Receptor for advanced glycation endproducts (RAGE) is a key regulator of oval cell activation and inflammation-associated liver carcinogenesis in mice. Hepatology 2013; 58:363-73. [PMID: 23504974 DOI: 10.1002/hep.26395] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Revised: 03/12/2013] [Accepted: 03/21/2013] [Indexed: 12/12/2022]
Abstract
UNLABELLED The receptor for advanced glycation endproducts (RAGE) is a multiligand receptor and member of the immunoglobulin superfamily. RAGE is mainly involved in tissue damage and chronic inflammatory disorders, sustaining the inflammatory response upon engagement with damage-associated molecular pattern molecules (DAMPs) such as S100 proteins and high-mobility group box 1 (HMGB1). Enhanced expression of RAGE and its ligands has been demonstrated in distinct tumors and several studies support its crucial role in tumor progression and metastasis by still unknown mechanisms. Here we show that RAGE supports hepatocellular carcinoma (HCC) formation in the Mdr2(-/-) mouse model, a prototype model of inflammation-driven HCC formation, which mimics the human pathology. Mdr2(-/-) Rage(-/-) (dKO) mice developed smaller and fewer HCCs than Mdr2(-/-) mice. Interestingly, although in preneoplastic Mdr2(-/-) livers RAGE ablation did not affect the onset of inflammation, premalignant dKO livers showed reduced liver damage and fibrosis, in association with decreased oval cell activation. Oval cells expressed high RAGE levels and displayed reduced proliferation upon RAGE silencing. Moreover, stimulation of oval cells with HMGB1 promoted an ERK1/2-Cyclin D1-dependent oval cell proliferation in vitro. Finally, genetic and pharmacologic blockade of RAGE signaling impaired oval cell activation in an independent mouse model of oval cell activation, the choline deficient ethionine-supplemented dietary regime. CONCLUSION Our data identified a novel function of RAGE in regulating oval cell activation and tumor development in inflammation-associated liver carcinogenesis.
Collapse
Affiliation(s)
- Tobias Pusterla
- Division of Signal Transduction and Growth Control, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Yu M, Zhou X, Niu L, Lin G, Huang J, Zhou W, Gan H, Wang J, Jiang X, Yin B, Li Z. Targeting transmembrane TNF-α suppresses breast cancer growth. Cancer Res 2013; 73:4061-74. [PMID: 23794706 DOI: 10.1158/0008-5472.can-12-3946] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
TNF antagonists may offer therapeutic potential in solid tumors, but patients who have high serum levels of TNF-α fail to respond to infliximab, suggesting consumption of the circulating antibody and loss of transmembrane TNF-α (tmTNF-α) on tumors by ectodomain shedding. Addressing this possibility, we developed a monoclonal antibody (mAb) that binds both full-length tmTNF-α and its N-terminal truncated fragment on the membrane after tmTNF-α processing but does not cross-react with soluble TNF-α. We documented high levels of tmTNF-α expression in primary breast cancers, lower levels in atypical hyperplasia or hyperplasia, but undetectable levels in normal breast tissue, consistent with the notion that tmTNF-α is a potential therapeutic target. Evaluations in vitro and in vivo further supported this assertion. tmTNF-α mAb triggered antibody-dependent cell-mediated cytotoxicity against tmTNF-α-expressing cells but not to tmTNF-α-negative cells. In tumor-bearing mice, tmTNF-α mAb delayed tumor growth, eliciting complete tumor regressions in some mice. Moreover, tmTNF-α mAb inhibited metastasis and expression of CD44v6, a prometastatic molecule. However, the antibody did not activate tmTNF-α-mediated reverse signaling, which facilitates tumor survival and resistance to apoptosis, but instead inhibited NF-κB activation and Bcl-2 expression by decreasing tmTNF-α-positive cells. Overall, our results established that tmTNF-α mAb exerts effective antitumor activities and offers a promising candidate to treat tmTNF-α-positive tumors, particularly in patients that are nonresponders to TNF antagonists.
Collapse
Affiliation(s)
- Mingxia Yu
- Department of Immunology, Tongji Medical College, Wuhan University, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Minicis SD, Kisseleva T, Francis H, Baroni GS, Benedetti A, Brenner D, Alvaro D, Alpini G, Marzioni M. Liver carcinogenesis: rodent models of hepatocarcinoma and cholangiocarcinoma. Dig Liver Dis 2013; 45. [PMID: 23177172 PMCID: PMC3716909 DOI: 10.1016/j.dld.2012.10.008] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Hepatocellular carcinoma and cholangiocarcinoma are primary liver cancers, both represent a growing challenge for clinicians due to their increasing morbidity and mortality. In the last few years a number of in vivo models of hepatocellular carcinoma and cholangiocarcinoma have been developed. The study of these models is providing a significant contribution in unveiling the pathophysiology of primary liver malignancies. They are also fundamental tools to evaluate newly designed molecules to be tested as new potential therapeutic agents in a pre-clinical set. Technical aspects of each model are critical steps, and they should always be considered in order to appropriately interpret the findings of a study or its planning. The purpose of this review is to describe the technical and experimental features of the most significant rodent models, highlighting similarities or differences between the corresponding human diseases. The first part is dedicated to the discussion of models of hepatocellular carcinoma, developed using toxic agents, or through dietary or genetic manipulations. In the second we will address models of cholangiocarcinoma developed in rats or mice by toxin administration, genetic manipulation and/or bile duct incannulation or surgery. Xenograft or syngenic models are also proposed.
Collapse
Affiliation(s)
- Samuele De Minicis
- Department of Gastroenterology, Università Politecnica delle Marche, Ancona, Italy
| | - Tatiana Kisseleva
- Division of Gastroenterology, Department of Medicine, University of California San Diego, School of Medicine, CA, United States
| | - Heather Francis
- Division Research, Central Texas Veterans Health Care System, Scott & White Digestive Disease Research Center, Department of Medicine, Division Gastroenterology, Scott & White Hospital and Texas A&M Health Science Center, College of Medicine, Temple, TX, United States
| | | | - Antonio Benedetti
- Department of Gastroenterology, Università Politecnica delle Marche, Ancona, Italy
| | - David Brenner
- Division of Gastroenterology, Department of Medicine, University of California San Diego, School of Medicine, CA, United States
| | - Domenico Alvaro
- Division of Gastroenterology, Polo Pontino, Università degli Studi “La Sapienza”, Rome, Italy
| | - Gianfranco Alpini
- Division of Gastroenterology, Department of Medicine, University of California San Diego, School of Medicine, CA, United States,Co-corresponding author. Tel.: +1 254 743 1041/1044; fax: +1 254 743 0378/0555. (M. Marzioni)
| | - Marco Marzioni
- Department of Gastroenterology, Università Politecnica delle Marche, Ancona, Italy,Corresponding author at: Department of Gastroenterology, Università Politecnica delle Marche, Nuovo Polo Didattico, III Piano, Via Tronto 10, 60020 Ancona, Italy. Tel.: +39 0712206043; fax: +39 0712206044
| |
Collapse
|
81
|
Ichinohe N, Tanimizu N, Ooe H, Nakamura Y, Mizuguchi T, Kon J, Hirata K, Mitaka T. Differentiation capacity of hepatic stem/progenitor cells isolated from D-galactosamine-treated rat livers. Hepatology 2013; 57:1192-1202. [PMID: 22991225 DOI: 10.1002/hep.26084] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Accepted: 09/08/2012] [Indexed: 12/07/2022]
Abstract
UNLABELLED Oval cells and small hepatocytes (SHs) are known to be hepatic stem and progenitor cells. Although oval cells are believed to differentiate into mature hepatocytes (MHs) through SHs, the details of their differentiation process are not well understood. Furthermore, it is not certain whether the induced cells possess fully mature functions as MHs. In the present experiment, we used Thy1 and CD44 to isolate oval and progenitor cells, respectively, from D-galactosamine-treated rat livers. Epidermal growth factor, basic fibroblast growth factor, or hepatocyte growth factor could trigger the hepatocytic differentiation of sorted Thy1(+) cells to form epithelial cell colonies, and the combination of the factors stimulated the emergence and expansion of the colonies. Cells in the Thy1(+) -derived colonies grew more slowly than those in the CD44(+) -derived ones in vitro and in vivo and the degree of their hepatocytic differentiation increased with CD44 expression. Although the induced hepatocytes derived from Thy1(+) and CD44(+) cells showed similar morphology to MHs and formed organoids from the colonies similar to those from SHs, many hepatic differentiated functions of the induced hepatocytes were less well performed than those of mature SHs derived from the healthy liver. The gene expression of cytochrome P450 1A2, tryptophan 2,3-dioxygenase, and carbamoylphosphate synthetase I was lower in the induced hepatocytes than in mature SHs. In addition, the protein expression of CCAAT/enhancer-binding protein alpha and bile canalicular formation could not reach the levels of production of mature SHs. CONCLUSION The results suggest that, although Thy1(+) and CD44(+) cells are able to differentiate into hepatocytes, the degree of maturation of the induced hepatocytes may not be equal to that of healthy resident hepatocytes. (HEPATOLOGY 2013).
Collapse
Affiliation(s)
- Norihisa Ichinohe
- Department of Tissue Development and Regeneration, the Research Institute for Frontier Medicine, Sapporo, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
82
|
Davoodi SH, Malek-Shahabi T, Malekshahi-Moghadam A, Shahbazi R, Esmaeili S. Obesity as an important risk factor for certain types of cancer. IRANIAN JOURNAL OF CANCER PREVENTION 2013; 6:186-94. [PMID: 25250133 PMCID: PMC4142931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 09/28/2013] [Indexed: 11/10/2022]
Abstract
Cancer could be described as the uncontrolled and unrestricted growth of malignant cells in any place of the body. It is a multifactorial disease which either heredity or environmental factors (such as nutrition, physical inactivity, alcohol, obesity, exposure to sun, environmental pollutants, infections) chip in incidence of cancer. In recent years, several researchers have focused on obesity as a potent cancer risk factor. Scientificevidences have suggested that obesity has associated with increased risk for a plenty of different types of cancer. The evidences are the most consistent for endometrial cancer, breast cancer between the postmenopausal women, and renal cell cancer. More contradictoryresults have reported about the colorectal, prostate, and pancreatic cancer. Although numerous studies have done according to the obesity and cancer relation or joint, but the molecular mechanisms in which obesity could increase the risks of cancer, have been poorly understood.
Collapse
Affiliation(s)
- Sayed Hossain Davoodi
- Dept. of Clinical Nutrition and Dietetics, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Sciences, Food Science and Technology, Shahid Beheshti University of Medical Sciences,Tehran, Iran
| | | | | | - Roghieh Shahbazi
- Dept. of Clinical Nutrition and Dietetics, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Sciences, Food Science and Technology, Shahid Beheshti University of Medical Sciences,Tehran, Iran
| | - Saeideh Esmaeili
- Dept. of Food Technology Research, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran,Corresponding Author: Saeideh Esmaeili, MSc
Tel: (+98) 21 22 96 91 77
| |
Collapse
|
83
|
Wang K, Lin B. Pathophysiological Significance of Hepatic Apoptosis. ISRN HEPATOLOGY 2012; 2013:740149. [PMID: 27335822 PMCID: PMC4890876 DOI: 10.1155/2013/740149] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2012] [Accepted: 12/13/2012] [Indexed: 12/19/2022]
Abstract
Apoptosis is a classical pathological feature in liver diseases caused by various etiological factors such as drugs, viruses, alcohol, and cholestasis. Hepatic apoptosis and its deleterious effects exacerbate liver function as well as involvement in fibrosis/cirrhosis and carcinogenesis. An imbalance between apoptotic and antiapoptotic capabilities is a prominent characteristic of liver injury. The regulation of apoptosis and antiapoptosis can be a pivotal step in the treatment of liver diseases.
Collapse
Affiliation(s)
- Kewei Wang
- Departments of Surgery and Pediatrics, University of Illinois College of Medicine at Peoria, Peoria, IL 61605, USA
| | - Bingliang Lin
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| |
Collapse
|
84
|
Ye Q, Lian F, Chavez PRG, Chung J, Ling W, Qin H, Seitz HK, Wang XD. Cytochrome P450 2E1 inhibition prevents hepatic carcinogenesis induced by diethylnitrosamine in alcohol-fed rats. Hepatobiliary Surg Nutr 2012; 1:5-18. [PMID: 23543859 DOI: 10.3978/j.issn.2304-3881.2012.11.05] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Chronic alcohol ingestion increases hepatic cytochrome P450 2E1 (CYP2E1), which is associated with hepatocarcinogenesis. We investigated whether treatment with chlormethiazole (CMZ), a CYP2E1 inhibitor, protects against alcohol-associated hepatic carcinogenesis in rats. Rats were fed either an ethanol liquid diet or a non-ethanol liquid diet, with or without CMZ for one and ten months. A single intraperitoneal injection of diethylnitrosamine (DEN, 20 mg/kg) was given to initiate hepatic carcinogenesis. CYP2E1 expression, inflammatory proteins, cell proliferation, protein-bound 4-HNE, etheno-DNA adducts, 8-hydroxy-2'-deoxyguanosine (8-OHdG), retinoid concentrations, and hepatic carcinogenesis were examined. Ethanol feeding for 1 month with DEN resulted in significantly increased hepatic CYP2E1 levels and increased nuclear accumulation of NF-κB protein and TNF-α expression, which were associated with increased cyclin D1 expression and p-GST positive altered hepatic foci. All of these changes induced by ethanol feeding were significantly inhibited by the one month CMZ treatment. At 10-months of treatment, hepatocellular adenomas were detected in ethanol-fed rats only, but neither in control rats nor in animals receiving ethanol and CMZ. The 8-OHdG formation was found to be significantly increased in ethanol fed animals and normalized with CMZ treatment. In addition, alcohol-reduced hepatic retinol and retinoic acid concentrations were restored by CMZ treatment to normal levels in the rats at 10 months of treatment. These data demonstrate that the inhibition of ethanol-induced CYP2E1 as a key pathogenic factor can counteract the tumor-promoting action of ethanol by decreasing TNF-α expression, NF-κB activation, and oxidative DNA damage as well as restoring normal hepatic levels of retinoic acid in DEN-treated rats.
Collapse
Affiliation(s)
- Qinyuan Ye
- Nutrition and Cancer Biology Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA 02111, USA ; School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | | | | | | | | | | | | | | |
Collapse
|
85
|
Generation, characterization and potential therapeutic applications of mature and functional hepatocytes from stem cells. J Cell Physiol 2012; 228:298-305. [DOI: 10.1002/jcp.24150] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
86
|
Qiu B, Wang X, Zhang P, Shi C, Zhang J, Qiu W, Wang W, Li D. Association of TNF-α promoter polymorphisms with the outcome of persistent HBV infection in a northeast Chinese Han population. Acta Biochim Biophys Sin (Shanghai) 2012; 44:712-8. [PMID: 22695741 DOI: 10.1093/abbs/gms046] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Tumor necrosis factor-α (TNF-α) plays an important role in the pathogenesis and clinical outcome of chronic hepatitis B virus (HBV) infection. The objective of this study was to evaluate the relationship between functional polymorphisms of TNF-α and different outcomes of persistent HBV infection in a northeast Chinese Han population. Here 189 HBV spontaneously recovered subjects (SR), 571 HBV-infected patients including 180 chronic hepatitis B (CHB), 196 liver cirrhosis (LC), and 195 hepatocellular carcinoma (HCC) individuals were enrolled in this study. All the samples were genotyped for TNF-α -857C/T and -863C/A using the polymerase chain reaction-restriction fragment length polymorphism method. The frequency of -857CC genotype was significantly higher in CHB and LC individuals compared with that of SR subjects (P= 0.03, OR = 1.57, 95% CI 1.04-2.39 and P= 0.03, OR = 1.57, 95% CI 1.04-2.35, respectively). A significant difference in the distribution of the allele -857C was observed for both CHB vs. SR (P= 0.01, OR = 1.52, 95% CI 1.08-2.13) and LC vs. SR (P= 0.02, OR = 1.47, 95% CI 1.06-2.04) cohorts. In addition, the frequency of -863AA genotype was significantly higher in CHB and LC patients than that of SR subjects (P= 0.01, OR = 3.90, 95% CI 1.35-11.23 and P= 0.01, OR = 3.83, 95% CI 1.34-10.96, respectively), and allele -863A frequency was significantly more common in CHB, LC, and HCC cohorts than that of SR controls (P= 0.004, OR = 1.72, 95% CI 1.19-2.50; P= 0.001, OR = 1.81, 95% CI 1.26-2.61 and P= 0.001, OR = 1.90, 95% CI 1.33-2.73, respectively). Our data also revealed that haplotype CA was strongly associated with persistent HBV infection. These results suggest an association between the TNF-α promoter variants and different outcomes of persistent HBV infection in the studied population.
Collapse
Affiliation(s)
- Bing Qiu
- Department of Gastroenterology, Heilongjiang Province Hospital, Harbin 150036, China.
| | | | | | | | | | | | | | | |
Collapse
|
87
|
Feng D, Kong X, Weng H, Park O, Wang H, Dooley S, Gershwin ME, Gao B. Interleukin-22 promotes proliferation of liver stem/progenitor cells in mice and patients with chronic hepatitis B virus infection. Gastroenterology 2012; 143:188-98.e7. [PMID: 22484119 PMCID: PMC3384505 DOI: 10.1053/j.gastro.2012.03.044] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Revised: 03/06/2012] [Accepted: 03/29/2012] [Indexed: 12/19/2022]
Abstract
BACKGROUND & AIMS Proliferation of liver stem/progenitor cells (LPCs), which can differentiate into hepatocytes or biliary epithelial cells, is often observed in chronically inflamed regions of liver in patients. We investigated how inflammation might promote proliferation of LPCs. METHODS We examined the role of interleukin (IL)-22, a survival factor for hepatocytes, on proliferation of LPCs in patients with chronic hepatitis B virus (HBV) infection and in mice. Proliferation of LPCs in mice was induced by feeding a diet that contained 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC). RESULTS Hepatic expression of IL-22 was increased in patients with HBV and correlated with the grade of inflammation and proliferation of LPCs. Mice on the DDC diet that overexpressed an IL-22 transgene specifically in liver (IL-22TG), or that were infected with an IL-22-expressing adenovirus, had increased proliferation of LPCs. Signal transducer and activator of transcription (STAT) 3, a component of the IL-22 signaling pathway, was activated in LPCs isolated from DDC-fed IL-22TG mice. Deletion of STAT3 from livers of IL-22TG mice reduced proliferation of LPCs. In addition, the receptors IL-22R1 and IL-10R2 were detected on epithelial cell adhesion molecule(+)CD45(-) LPCs isolated from DDC-fed wild-type mice. Culture of these cells with IL-22 activated STAT3 and led to cell proliferation, but IL-22 had no effect on proliferation of STAT3-deficient EpCAM(+)CD45(-) LPCs. IL-22 also activated STAT3 and promoted proliferation of cultured BMOL cells (a mouse LPC line). CONCLUSIONS In livers of mice and patients with chronic HBV infection, inflammatory cells produce IL-22, which promotes proliferation of LPCs via STAT3. These findings link inflammation with proliferation of LPCs in patients with HBV infection.
Collapse
Affiliation(s)
- Dechun Feng
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Xiaoni Kong
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Honglei Weng
- Molecular Hepatology, II. Medical Clinic, Faculty of Medicine Mannheim at Heidelberg University, Germany
| | - Ogyi Park
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Hua Wang
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Steven Dooley
- Molecular Hepatology, II. Medical Clinic, Faculty of Medicine Mannheim at Heidelberg University, Germany
| | - M. Eric Gershwin
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis School of Medicine, Davis, CA, USA
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
88
|
TNF-inducible expression of lymphotoxin-β in hepatic cells: an essential role for NF-κB and Ets1 transcription factors. Cytokine 2012; 60:498-504. [PMID: 22742857 DOI: 10.1016/j.cyto.2012.05.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Revised: 05/30/2012] [Accepted: 05/31/2012] [Indexed: 11/21/2022]
Abstract
As TNF is one of the earliest signals that can be detected in the leukocyte-derived inflammatory cascade which drives subsequent cytokine production, we are interested in determining whether TNF is one of the initiating factors controlling liver remodeling and regeneration following chronic liver damage. One of the early responses is the expression of lymphotoxin-β by hepatic progenitor oval cells. The aim of this study was to determine whether hepatic expression of LT-β was controlled by TNF and to understand the basis of this regulation. We previously showed that LT-β expression is transcriptionally controlled via the TNF-induced, inflammatory NF-κB pathway in T lymphocytes. Here we show that TNF is able to upregulate LT-β expression in hepatic cells at the transcriptional level by the binding of NF-κB p50/p65 heterodimers and Ets1 to their respective sites in the LT-β promoter.
Collapse
|
89
|
Ho MY, Tang SJ, Chuang MJ, Cha TL, Li JY, Sun GH, Sun KH. TNF-α induces epithelial-mesenchymal transition of renal cell carcinoma cells via a GSK3β-dependent mechanism. Mol Cancer Res 2012; 10:1109-19. [PMID: 22707636 DOI: 10.1158/1541-7786.mcr-12-0160] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
TNF-α is a cytokine with antitumorigenic property. In contrast, low dose, chronic TNF-α production by tumor cells or stromal cells may promote tumor growth and metastasis. Serum levels of TNF-α are significantly elevated in renal cell carcinoma (RCC) patients. Here, we showed that TNF-α induced epithelial-mesenchymal transition (EMT) and promoted tumorigenicity of RCC by repressing E-cadherin, upregulating vimentin, activating MMP9, and invasion activities. In addition, TNF-α treatment inhibited glycogen synthase kinase 3β (GSK-3β) activity through serine-9 phosphorylation mediated by the phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) pathway in RCC cells. Inhibition of PI3K/AKT by LY294002 reactivated GSK-3β and suppressed the TNF-α-induced EMT of RCC cells. Inactivation of GSK-3β by LiCl significantly increased MMP9 activity and EMT of RCC cells. Activation of GSK-3β by transduction of constitutively active GSK-3β into RCC cells suppressed TNF-α-mediated anchorage-independent growth in soft agar and tumorigenicity in nude mice. Overexpression of a kinase-deficient GSK-3β, in contrast, potentiated EMT, anchorage-independent growth and drastically enhanced tumorigenicity in vivo. Most importantly, a 15-fold inactivation of GSK-3β activity, 3-fold decrease of E-cadherin, and 2-fold increase of vimentin were observed in human RCC tumor tissues. These results indicated that inactivation of GSK-3β plays a pivotal role in the TNF-α-mediated tumorigenesis of RCC.
Collapse
Affiliation(s)
- Ming-Yi Ho
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
90
|
Radwan MI, Pasha HF, Mohamed RH, Hussien HIM, El-Khshab MN. Influence of transforming growth factor-β1 and tumor necrosis factor-α genes polymorphisms on the development of cirrhosis and hepatocellular carcinoma in chronic hepatitis C patients. Cytokine 2012; 60:271-6. [PMID: 22682513 DOI: 10.1016/j.cyto.2012.05.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Revised: 05/13/2012] [Accepted: 05/14/2012] [Indexed: 02/07/2023]
Abstract
BACKGROUND Host genetic factors may affect clinical outcomes of hepatitis C virus (HCV) infection; however, the possible mechanisms remain largely unknown. This study aimed to evaluate transforming growth factor-β1 (TGF-β1)-509 and tumor necrosis factor-α (TNF-α)-308 genes polymorphisms as a risk factors for cirrhosis and hepatocellular carcinoma (HCC) in chronic hepatitis C patients. MATERIALS AND METHODS Two hundred and eighty HCV patients (152 patients with cirrhosis, 128 patients with HCC) and 160 controls were enrolled in the study. Polymorphisms of TGF-β1-509 and TNF-α-308 gene were determined using polymerase chain reaction restriction fragment length polymorphism (PCR-RFLP). Serum TGF-β1 and TNF-α were determined using ELISA. RESULTS TGF-β1-509 TT, TNF-α-308 AA and GA genotypes frequencies were significantly increased in cirrhotic and HCC groups. Serum TGF-β1 and TNF-α level were significantly increased in TGF-β1-509 TT and TNF-α-308 AA genotypes respectively. CONCLUSION TGF-β1-509 and TNF-α-308 genes polymorphisms are associated with risk of liver cirrhosis and HCC in patients with chronic HCV infection.
Collapse
Affiliation(s)
- Mohamed I Radwan
- Tropical Medicine Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | | | | | | | | |
Collapse
|
91
|
Shiozaki A, Bai XH, Shen-Tu G, Moodley S, Takeshita H, Fung SY, Wang Y, Keshavjee S, Liu M. Claudin 1 mediates TNFα-induced gene expression and cell migration in human lung carcinoma cells. PLoS One 2012; 7:e38049. [PMID: 22675434 PMCID: PMC3365005 DOI: 10.1371/journal.pone.0038049] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2011] [Accepted: 05/02/2012] [Indexed: 12/26/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is an important mechanism in carcinogenesis. To determine the mechanisms that are involved in the regulation of EMT, it is crucial to develop new biomarkers and therapeutic targets towards cancers. In this study, when TGFβ1 and TNFα were used to induce EMT in human lung carcinoma A549 cells, we found an increase in an epithelial cell tight junction marker, Claudin 1. We further identified that it was the TNFα and not the TGFβ1 that induced the fibroblast-like morphology changes. TNFα also caused the increase in Claudin-1 gene expression and protein levels in Triton X-100 soluble cytoplasm fraction. Down-regulation of Claudin-1, using small interfering RNA (siRNA), inhibited 75% of TNFα-induced gene expression changes. Claudin-1 siRNA effectively blocked TNFα-induced molecular functional networks related to inflammation and cell movement. Claudin-1 siRNA was able to significantly reduce TNF-enhanced cell migration and fibroblast-like morphology. Furthermore, over expression of Claudin 1 with a Claudin 1-pcDNA3.1/V5-His vector enhanced cell migration. In conclusion, these observations indicate that Claudin 1 acts as a critical signal mediator in TNFα-induced gene expression and cell migration in human lung cancer cells. Further analyses of these cellular processes may be helpful in developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Atsushi Shiozaki
- Latner Thoracic Surgery Research Laboratories, University Health Network Toronto General Research Institute, Toronto, Canada
| | - Xiao-hui Bai
- Latner Thoracic Surgery Research Laboratories, University Health Network Toronto General Research Institute, Toronto, Canada
| | - Grace Shen-Tu
- Latner Thoracic Surgery Research Laboratories, University Health Network Toronto General Research Institute, Toronto, Canada
| | - Serisha Moodley
- Latner Thoracic Surgery Research Laboratories, University Health Network Toronto General Research Institute, Toronto, Canada
| | - Hiroki Takeshita
- Latner Thoracic Surgery Research Laboratories, University Health Network Toronto General Research Institute, Toronto, Canada
| | - Shan-Yu Fung
- Latner Thoracic Surgery Research Laboratories, University Health Network Toronto General Research Institute, Toronto, Canada
| | - Yingchun Wang
- Latner Thoracic Surgery Research Laboratories, University Health Network Toronto General Research Institute, Toronto, Canada
| | - Shaf Keshavjee
- Latner Thoracic Surgery Research Laboratories, University Health Network Toronto General Research Institute, Toronto, Canada
- Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Mingyao Liu
- Latner Thoracic Surgery Research Laboratories, University Health Network Toronto General Research Institute, Toronto, Canada
- Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Canada
- * E-mail:
| |
Collapse
|
92
|
van der Weyden L, Adams DJ. Using mice to unveil the genetics of cancer resistance. Biochim Biophys Acta Rev Cancer 2012; 1826:312-30. [PMID: 22613679 DOI: 10.1016/j.bbcan.2012.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Revised: 05/10/2012] [Accepted: 05/13/2012] [Indexed: 11/28/2022]
Abstract
In the UK, four in ten people will develop some form of cancer during their lifetime, with an individual's relative risk depending on many factors, including age, lifestyle and genetic make-up. Much research has gone into identifying the genes that are mutated in tumorigenesis with the overwhelming majority of genetically-modified (GM) mice in cancer research showing accelerated tumorigenesis or recapitulating key aspects of the tumorigenic process. Yet if six out of ten people will not develop some form of cancer during their lifetime, together with the fact that some cancer patients experience spontaneous regression/remission, it suggests there are ways of 'resisting' cancer. Indeed, there are wildtype, spontaneously-arising mutants and GM mice that show some form of 'resistance' to cancer. Identification of mice with increased resistance to cancer is a novel aspect of cancer research that is important in terms of providing both chemopreventative and therapeutic options. In this review we describe the different mouse lines that display a 'cancer resistance' phenotype and discuss the molecular basis of their resistance.
Collapse
Affiliation(s)
- Louise van der Weyden
- Experimental Cancer Genetics, Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK.
| | | |
Collapse
|
93
|
Xiang S, Dong HH, Liang HF, He SQ, Zhang W, Li CH, Zhang BX, Zhang BH, Jing K, Tomlinson S, van Rooijen N, Jiang L, Cianflone K, Chen XP. Oval cell response is attenuated by depletion of liver resident macrophages in the 2-AAF/partial hepatectomy rat. PLoS One 2012; 7:e35180. [PMID: 22514719 PMCID: PMC3325996 DOI: 10.1371/journal.pone.0035180] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 03/09/2012] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND/AIMS Macrophages are known to play an important role in hepatocyte mediated liver regeneration by secreting inflammatory mediators. However, there is little information available on the role of resident macrophages in oval cell mediated liver regeneration. In the present study we aimed to investigate the role of macrophages in oval cell expansion induced by 2-acetylaminofluorene/partial hepatectomy (2-AAF/PH) in rats. METHODOLOGY/PRINCIPAL FINDINGS We depleted macrophages in the liver of 2-AAF/PH treated rats by injecting liposome encapsulated clodronate 48 hours before PH. Regeneration of remnant liver mass, as well as proliferation and differentiation of oval cells were measured. We found that macrophage-depleted rats suffered higher mortality and liver transaminase levels. We also showed that depletion of macrophages yielded a significant decrease of EPCAM and PCK positive oval cells in immunohistochemical stained liver sections 9 days after PH. Meanwhile, oval cell differentiation was also attenuated as a result of macrophage depletion, as large foci of small basophilic hepatocytes were observed by day 9 following hepatectomy in control rats whereas they were almost absent in macrophage depleted rats. Accordingly, real-time polymerase chain reaction analysis showed lower expression of albumin mRNA in macrophage depleted livers. Then we assessed whether macrophage depletion may affect hepatic production of stimulating cytokines for liver regeneration. We showed that macrophage-depletion significantly inhibited hepatic expression of tumor necrosis factor-α and interleukin-6, along with a lack of signal transducer and activator of transcription 3 phosphorylation during the early period following hepatectomy. CONCLUSIONS These data indicate that macrophages play an important role in oval cell mediated liver regeneration in the 2-AAF/PH model.
Collapse
Affiliation(s)
- Shuai Xiang
- Hepatic Surgery Centre, Huazhong University of Science and Technology, Tongji Hospital, Wuhan, China
| | - Han-Hua Dong
- Hepatic Surgery Centre, Huazhong University of Science and Technology, Tongji Hospital, Wuhan, China
| | - Hui-Fang Liang
- Hepatic Surgery Centre, Huazhong University of Science and Technology, Tongji Hospital, Wuhan, China
| | - Song-Qing He
- Department of Hepatobiliary Surgery, Guilin Medical University, Affiliated Hospital, Guilin, China
| | - Wei Zhang
- Department of Hepatobiliary Surgery, Guilin Medical University, Affiliated Hospital, Guilin, China
| | - Chang-Hai Li
- Hepatic Surgery Centre, Huazhong University of Science and Technology, Tongji Hospital, Wuhan, China
| | - Bi-Xiang Zhang
- Department of Hepatobiliary Surgery, Guilin Medical University, Affiliated Hospital, Guilin, China
| | - Bin-Hao Zhang
- Department of Hepatobiliary Surgery, Guilin Medical University, Affiliated Hospital, Guilin, China
| | - Kai Jing
- Department of Hepatobiliary Surgery, Guilin Medical University, Affiliated Hospital, Guilin, China
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Darby Children's Research Institute, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Nico van Rooijen
- Department of Molecular Cell Biology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | - Li Jiang
- Hepatic Surgery Centre, Huazhong University of Science and Technology, Tongji Hospital, Wuhan, China
| | - Katherine Cianflone
- Centre de Recherche Institut Universitaire de Cardiologie et de Pneumologie de Quebec, Université Laval, Quebec, Canada
| | - Xiao-Ping Chen
- Hepatic Surgery Centre, Huazhong University of Science and Technology, Tongji Hospital, Wuhan, China
- * E-mail:
| |
Collapse
|
94
|
Martin M, Herceg Z. From hepatitis to hepatocellular carcinoma: a proposed model for cross-talk between inflammation and epigenetic mechanisms. Genome Med 2012; 4:8. [PMID: 22293089 PMCID: PMC3334556 DOI: 10.1186/gm307] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Inflammation represents the body's natural response to tissue damage; however, chronic inflammation may activate cell proliferation and induce deregulation of cell death in affected tissues. Chronic inflammation is an important factor in the development of hepatocellular carcinoma (HCC), although the precise underlying mechanism remains unknown. Epigenetic events, which are considered key mechanisms in the regulation of gene activity states, are also commonly deregulated in HCC. Here, we review the evidence that chronic inflammation might deregulate epigenetic processes, thus promoting oncogenic transformation, and we propose a working hypothesis that epigenetic deregulation is an underlying mechanism by which inflammation might promote HCC development. In this scenario, different components of the inflammatory response might directly and indirectly induce changes in epigenetic machineries ('epigenetic switch'), including those involved in setting and propagating normal patterns of DNA methylation, histone modifications and non-coding RNAs in hepatocytes. We discuss the possibility that self-reinforcing cross-talk between inflammation and epigenetic mechanisms might amplify inflammatory signals and maintain a chronic state of inflammation culminating in cancer development. The potential role of inflammation-epigenome interactions in the emergence and maintenance of cancer stem cells is also discussed.
Collapse
Affiliation(s)
- Marion Martin
- Epigenetics Group, International Agency for Research on Cancer (IARC), 150 Cours Albert Thomas, 69372 Lyon CEDEX 08, France
| | - Zdenko Herceg
- Epigenetics Group, International Agency for Research on Cancer (IARC), 150 Cours Albert Thomas, 69372 Lyon CEDEX 08, France
| |
Collapse
|
95
|
Aggarwal BB, Gupta SC, Kim JH. Historical perspectives on tumor necrosis factor and its superfamily: 25 years later, a golden journey. Blood 2012; 119:651-65. [PMID: 22053109 PMCID: PMC3265196 DOI: 10.1182/blood-2011-04-325225] [Citation(s) in RCA: 581] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Accepted: 10/31/2011] [Indexed: 12/15/2022] Open
Abstract
Although activity that induced tumor regression was observed and termed tumor necrosis factor (TNF) as early as the 1960s, the true identity of TNF was not clear until 1984, when Aggarwal and coworkers reported, for the first time, the isolation of 2 cytotoxic factors: one, derived from macrophages (molecular mass 17 kDa), was named TNF, and the second, derived from lymphocytes (20 kDa), was named lymphotoxin. Because the 2 cytotoxic factors exhibited 50% amino acid sequence homology and bound to the same receptor, they came to be called TNF-α and TNF-β. Identification of the protein sequences led to cloning of their cDNA. Based on sequence homology to TNF-α, now a total of 19 members of the TNF superfamily have been identified, along with 29 interacting receptors, and several molecules that interact with the cytoplasmic domain of these receptors. The roles of the TNF superfamily in inflammation, apoptosis, proliferation, invasion, angiogenesis, metastasis, and morphogenesis have been documented. Their roles in immunologic, cardiovascular, neurologic, pulmonary, and metabolic diseases are becoming apparent. TNF superfamily members are active targets for drug development, as indicated by the recent approval and expanding market of TNF blockers used to treat rheumatoid arthritis, psoriasis, Crohns disease, and osteoporosis, with a total market of more than US $20 billion. As we learn more about this family, more therapeutics will probably emerge. In this review, we summarize the initial discovery of TNF-α, and the insights gained regarding the roles of this molecule and its related family members in normal physiology and disease.
Collapse
Affiliation(s)
- Bharat B Aggarwal
- Cytokine Research Laboratory, Department of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, 77054, USA.
| | | | | |
Collapse
|
96
|
Chobert MN, Couchie D, Fourcot A, Zafrani ES, Laperche Y, Mavier P, Brouillet A. Liver precursor cells increase hepatic fibrosis induced by chronic carbon tetrachloride intoxication in rats. J Transl Med 2012; 92:135-50. [PMID: 21946857 PMCID: PMC3425737 DOI: 10.1038/labinvest.2011.143] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hepatic fibrosis, the major complication of virtually all types of chronic liver damage, usually begins in portal areas, and its severity has been correlated to liver progenitor cells (LPC) expansion from periportal areas, even if the primary targets of injury are intralobular hepatocytes. The aim of this study was to determine the potential fibrogenic role of LPC, using a new experimental model in which rat liver fibrosis was induced by chronic carbon tetrachloride (CCl(4)) administration for 6 weeks, in combination with chronic acetylaminofluorene treatment (AAF), which promotes activation of LPC compartment. Treatment with CCl(4) alone caused a significant increase in serum transaminase activity as well as liver fibrosis initiating around central veins and leading to formation of incomplete centro-central septa with sparse fibrogenic cells expressing α-smooth muscle actin (αSMA). In AAF/CCl(4)-treated animals, the fibrogenic response was profoundly worsened, with formation of multiple porto-central bridging septa leading to cirrhosis, whereas hepatocellular necrosis and inflammation were similar to those observed in CCl(4)-treated animals. Enhanced fibrosis in AAF/CCl(4) group was accompanied by ductule forming LPC expanding from portal areas, αSMA-positive cells accumulation in the fibrotic areas and increased expression of hepatic collagen type 1, 3 and 4 mRNA. Moreover, CK19-positive LPC expressed the most potent fibrogenic cytokine transforming growth factor-β (TGFβ) without any expression of αSMA, desmin or fibroblast-specific protein-1, demonstrating that LPC did not undergo an epithelial-mesenchymal transition. In this new experimental model, LPC, by expressing TGFβ, contributed to the accumulation of αSMA-positive myofibroblasts in the ductular reaction leading to enhanced fibrosis but also to disease progression and to a fibrotic pattern similar to that observed in humans.
Collapse
Affiliation(s)
- Marie-Noële Chobert
- Institut Mondor de Recherche Biomédicale
INSERM : U955Université Paris XII - Paris Est Créteil Val-de-MarneIFR108 rue du général Sarrail 94010 Créteil, FR
| | - Dominique Couchie
- Institut Mondor de Recherche Biomédicale
INSERM : U955Université Paris XII - Paris Est Créteil Val-de-MarneIFR108 rue du général Sarrail 94010 Créteil, FR
| | - Agnès Fourcot
- UFR Médecine
Université Paris XII - Paris Est Créteil Val-de-MarneAvenue du Général de Gaulle 94010 Créteil Cedex, FR
| | - Elie-Serge Zafrani
- Institut Mondor de Recherche Biomédicale
INSERM : U955Université Paris XII - Paris Est Créteil Val-de-MarneIFR108 rue du général Sarrail 94010 Créteil, FR,Service d'anatomie et cytologie pathologiques [Mondor]
Assistance publique - Hôpitaux de Paris (AP-HP)Hôpital Henri MondorUniversité Paris XII - Paris Est Créteil Val-de-Marne51 Av Maréchal de Lattre de Tassigny, 94000 Créteil,FR
| | - Yannick Laperche
- Institut Mondor de Recherche Biomédicale
INSERM : U955Université Paris XII - Paris Est Créteil Val-de-MarneIFR108 rue du général Sarrail 94010 Créteil, FR
| | - Philippe Mavier
- UFR Médecine
Université Paris XII - Paris Est Créteil Val-de-MarneAvenue du Général de Gaulle 94010 Créteil Cedex, FR
| | - Arthur Brouillet
- Institut Mondor de Recherche Biomédicale
INSERM : U955Université Paris XII - Paris Est Créteil Val-de-MarneIFR108 rue du général Sarrail 94010 Créteil, FR,* Correspondence should be adressed to: Arthur Brouillet
| |
Collapse
|
97
|
Abstract
The increasing incidence of obesity and its co-morbid conditions poses a great challenge to global health. In addition to cardiovascular disease and diabetes, epidemiological data demonstrate a link between obesity and multiple types of cancer. The molecular mechanisms underlying how obesity causes an increased risk of cancer are poorly understood. Obesity disrupts the dynamic role of the adipocyte in energy homeostasis, resulting in inflammation and alteration of adipokine (for example, leptin and adiponectin) signalling. Additionally, obesity causes secondary changes that are related to insulin signalling and lipid deregulation that may also foster cancer development. Understanding these molecular links may provide an avenue for preventive and therapeutic strategies to reduce cancer risk and mortality in an increasingly obese population.
Collapse
Affiliation(s)
- Melin J Khandekar
- Dana-Farber Cancer Institute and Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | |
Collapse
|
98
|
Kim JE, Son JE, Jang YJ, Lee DE, Kang NJ, Jung SK, Heo YS, Lee KW, Lee HJ. Luteolin, a novel natural inhibitor of tumor progression locus 2 serine/threonine kinase, inhibits tumor necrosis factor-alpha-induced cyclooxygenase-2 expression in JB6 mouse epidermis cells. J Pharmacol Exp Ther 2011; 338:1013-22. [PMID: 21705614 DOI: 10.1124/jpet.111.179200] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Targeting tumor necrosis factor (TNF)-α-mediated signal pathways may be a promising strategy for developing chemopreventive agents, because TNF-α-mediated cyclooxygenase (COX)-2 expression plays a key role in inflammation and carcinogenesis. Luteolin [2-(3,4-dihydroxyphenyl)-5,7-dihydroxy-4-chromenone] exerts anticarcinogenic effects, although little is known about the underlying molecular mechanisms and specific targets of this compound. In the present study, we found that luteolin inhibited TNF-α-induced COX-2 expression by down-regulating the transactivation of nuclear factor-κB and activator protein-1. Furthermore, luteolin inhibited TNF-α-induced phosphorylation of mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK) kinase 1/ERK/p90(RSK), mitogen-activated protein kinase kinase 4/c-Jun N-terminal kinase/c-Jun, and Akt/p70(S6K). However, it had no effect on the phosphorylation of p38. These effects of luteolin on TNF-α-mediated signaling pathways and COX-2 expression are similar to those achieved by blocking tumor progression locus 2 serine/threonine kinase (TPL2) using pharmacologic inhibitors and small interfering RNAs. Luteolin inhibited TPL2 activity in vitro and in TPL2 immunoprecipitation kinase assays by binding directly in an ATP-competitive manner. Overall, these results indicate that luteolin exerts potent chemopreventive activities, which primarily target TPL2.
Collapse
Affiliation(s)
- Jong-Eun Kim
- World Class University Biomodulation Program, Department of Agricultural Biotechnology, Center for Agricultural Biomaterials and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Jacobs PP, Sackstein R. CD44 and HCELL: preventing hematogenous metastasis at step 1. FEBS Lett 2011; 585:3148-58. [PMID: 21827751 DOI: 10.1016/j.febslet.2011.07.039] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2011] [Accepted: 07/21/2011] [Indexed: 12/23/2022]
Abstract
Despite great strides in our knowledge of the genetic and epigenetic changes underlying malignancy, we have limited information on the molecular basis of metastasis. Over 90% of cancer deaths are caused by spread of tumor cells from a primary site to distant organs and tissues, highlighting the pressing need to define the molecular effectors of cancer metastasis. Mounting evidence suggests that circulating tumor cells (CTCs) home to specific tissues by hijacking the normal leukocyte trafficking mechanisms. Cancer cells characteristically express CD44, and there is increasing evidence that hematopoietic cell E-/L-selectin ligand (HCELL), a sialofucosylated glycoform of CD44, serves as the major selectin ligand on cancer cells, allowing interaction of tumor cells with endothelium, leukocytes, and platelets. Here, we review the structural biology of CD44 and of HCELL, and present current data on the function of these molecules in mediating organ-specific homing/metastasis of CTCs.
Collapse
Affiliation(s)
- Pieter P Jacobs
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
100
|
Abstract
Mononuclear phagocytes often function as control switches of the immune system, securing the balance between pro- and anti-inflammatory reactions. For this purpose and depending on the activating stimuli, these cells can develop into different subsets: proinflammatory classically activated (M1) or anti-inflammatory alternatively activated (M2) macrophages. The expression of the nuclear peroxisome proliferator-activated receptors (PPARs) is regulated by M1- or M2-inducing stimuli, and these receptors are generally considered to counteract inflammatory M1 macrophages, while actively promoting M2 activation. This is of importance in a tumor context, where M1 are important initiators of inflammation-driven cancers. As a consequence, PPAR agonists are potentially usefull for inhibiting the early phases of tumorigenesis through their antagonistic effect on M1. In more established tumors, the macrophage phenotype is more diverse, making it more difficult to predict the outcome of PPAR agonism. Overall, in our view current knowledge provides a sound basis for the clinical evaluation of PPAR ligands as chemopreventive agents in chronic inflammation-associated cancer development, while cautioning against the unthoughtful application of these agents as cancer therapeutics.
Collapse
|