101
|
Bresnahan E, Lindblad KE, Ruiz de Galarreta M, Lujambio A. Mouse Models of Oncoimmunology in Hepatocellular Carcinoma. Clin Cancer Res 2020; 26:5276-5286. [PMID: 32327473 DOI: 10.1158/1078-0432.ccr-19-2923] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/10/2020] [Accepted: 04/20/2020] [Indexed: 12/13/2022]
Abstract
Liver cancer is the fourth leading cause of cancer-related mortality worldwide and incidence is on the rise. Hepatocellular carcinoma (HCC) is the most common form of liver cancer, with a complex etiology and limited treatment options. The standard-of-care treatment for patients with advanced HCC is sorafenib, a tyrosine kinase inhibitor that offers limited survival benefit. In the past years, therapeutic options for the treatment of advanced HCC have increased substantially, including additional multikinase inhibitors as well as immune checkpoint inhibitors. Nivolumab and pembrolizumab were approved in 2017 and 2018, respectively, as second-line treatment in advanced HCC. These drugs, both targeting the programmed death-1 pathway, demonstrate unprecedented results, with objective response rates of approximately 20%. However, the majority of patients do not respond, necessitating the identification of biomarkers of response and resistance to immunotherapy. With the recent success of immunotherapies in oncology, mouse models that better recapitulate the human disease and antitumor immune response are needed. This review lists ongoing clinical trials testing immunotherapy in HCC, briefly discusses the unique immunosuppressive environment of the liver, and then delves into the most applicable current murine model systems to study oncoimmunology within the context of HCC, including syngeneic, genetically engineered, and humanized models.
Collapse
Affiliation(s)
- Erin Bresnahan
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York.,Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Katherine E Lindblad
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York.,Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, New York
| | - Marina Ruiz de Galarreta
- Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Amaia Lujambio
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York. .,Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
102
|
Shizu R, Yoshinari K. Nuclear receptor CAR-mediated liver cancer and its species differences. Expert Opin Drug Metab Toxicol 2020; 16:343-351. [PMID: 32202166 DOI: 10.1080/17425255.2020.1746268] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction: The nuclear receptor CAR plays an important role in the regulation of hepatic responses to xenobiotic exposure, including the induction of hepatocyte proliferation and chemical carcinogenesis. Phenobarbital, a well-known liver cancer promoter, has been found to promote hepatocyte proliferation via CAR activation. However, the molecular mechanisms by which CAR induces liver carcinogenesis remain unknown. In addition, it is believed that CAR-mediated liver carcinogenesis shows a species difference; phenobarbital treatment induces hepatocyte proliferation and liver cancer in rodents but not in humans. However, the mechanisms are also unknown.Areas covered: Several reports indicate that the key oncogenic signaling pathways Wnt/β-catenin and Hippo/YAP are involved in CAR-mediated liver carcinogenesis. We introduce current data about the possible molecular mechanisms involved in CAR-mediated liver carcinogenesis and species differences by focusing on these two signaling pathways.Expert opinion: CAR may activate both the Wnt/β-catenin and Hippo/YAP signaling pathways. The synergistic activation of both signaling pathways seems to be important for CAR-mediated liver cancer development. Low homology between the ligand binding domains of human CAR and rodent CAR might cause species differences in the interactions with proteins that control the Wnt/β-catenin and Hippo/YAP pathways as well as liver cancer induction.
Collapse
Affiliation(s)
- Ryota Shizu
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Kouichi Yoshinari
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| |
Collapse
|
103
|
Tipanee J, Di Matteo M, Tulalamba W, Samara-Kuko E, Keirsse J, Van Ginderachter JA, Chuah MK, VandenDriessche T. Validation of miR-20a as a Tumor Suppressor Gene in Liver Carcinoma Using Hepatocyte-Specific Hyperactive piggyBac Transposons. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 19:1309-1329. [PMID: 32160703 PMCID: PMC7036702 DOI: 10.1016/j.omtn.2020.01.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 01/12/2020] [Accepted: 01/13/2020] [Indexed: 02/07/2023]
Abstract
We established a semi-high-throughput in vivo screening platform using hyperactive piggyBac (hyPB) transposons (designated as PB-miR) to identify microRNAs (miRs) that inhibit hepatocellular carcinoma (HCC) development in vivo, following miR overexpression in hepatocytes. PB-miRs encoding six different miRs from the miR-17-92 cluster and nine miRs from outside this cluster were transfected into mouse livers that were chemically induced to develop HCC. In this slow-onset HCC model, miR-20a significantly inhibited HCC. Next, we developed a more aggressive HCC model by overexpression of oncogenic Harvey rat sarcoma viral oncogene homolog (HRASG12V) and c-MYC oncogenes that accelerated HCC development after only 6 weeks. The tumor suppressor effect of miR-20a could be demonstrated even in this rapid-onset HRASG12V/c-MYC HCC model, consistent with significantly prolonged survival and decreased HCC tumor burden. Comprehensive RNA expression profiling of 95 selected genes typically associated with HCC development revealed differentially expressed genes and functional pathways that were associated with miR-20a-mediated HCC suppression. To our knowledge, this is the first study establishing a direct causal relationship between miR-20a overexpression and liver cancer inhibition in vivo. Moreover, these results demonstrate that hepatocyte-specific hyPB transposons are an efficient platform to screen and identify miRs that affect overall survival and HCC tumor regression.
Collapse
Affiliation(s)
- Jaitip Tipanee
- Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Mario Di Matteo
- Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium; Center for Molecular & Vascular Biology, Department of Cardiovascular Sciences, University of Leuven, 3000 Leuven, Belgium
| | - Warut Tulalamba
- Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Ermira Samara-Kuko
- Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Jiri Keirsse
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jo A Van Ginderachter
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Marinee Khim Chuah
- Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium; Center for Molecular & Vascular Biology, Department of Cardiovascular Sciences, University of Leuven, 3000 Leuven, Belgium.
| | - Thierry VandenDriessche
- Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium; Center for Molecular & Vascular Biology, Department of Cardiovascular Sciences, University of Leuven, 3000 Leuven, Belgium.
| |
Collapse
|
104
|
Enhanced DNA-repair capacity and resistance to chemically induced carcinogenesis upon deletion of the phosphatase regulator NIPP1. Oncogenesis 2020; 9:30. [PMID: 32123159 PMCID: PMC7051951 DOI: 10.1038/s41389-020-0214-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 02/12/2020] [Accepted: 02/13/2020] [Indexed: 11/16/2022] Open
Abstract
Nuclear Inhibitor of PP1 (NIPP1) is a conserved regulatory subunit of protein phosphatase PP1. The selective deletion of NIPP1 in mouse liver parenchymal cells or skin epidermal cells culminates in a late-onset hyperproliferation of a subset of resident progenitor cells. Although a hyperplastic phenotype is usually tumor promoting, we show here that the absence of NIPP1 conferred a strong resistance to chemically induced hepatocellular or skin carcinoma. The ablation of NIPP1 did not affect the metabolism of the administered mutagens (diethylnitrosamine or 7,12-dimethylbenz[a]anthracene), but reduced the conversion of mutagen-induced covalent DNA modifications into cancer-initiating mutations. This reduced sensitivity to mutagens correlated with an enhanced DNA-damage response and an augmented expression of rate-limiting DNA-repair proteins (MGMT in liver, XPD and XPG in skin), hinting at an increased DNA-repair capacity. Our data identify NIPP1 as a repressor of DNA repair and as a promising target for novel cancer prevention and treatment therapies.
Collapse
|
105
|
Novel patient-derived preclinical models of liver cancer. J Hepatol 2020; 72:239-249. [PMID: 31954489 DOI: 10.1016/j.jhep.2019.09.028] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/24/2019] [Accepted: 09/28/2019] [Indexed: 12/25/2022]
Abstract
Preclinical models of cancer based on the use of human cancer cell lines and mouse models have enabled discoveries that have been successfully translated into patients. And yet the majority of clinical trials fail, emphasising the urgent need to improve preclinical research to better interrogate the potential efficacy of each therapy and the patient population most likely to benefit. This is particularly important for liver malignancies, which lack highly efficient treatments and account for hundreds of thousands of deaths around the globe. Given the intricate network of genetic and environmental factors that contribute to liver cancer development and progression, the identification of new druggable targets will mainly depend on establishing preclinical models that mirror the complexity of features observed in patients. The development of new 3D cell culture systems, originating from cells/tissues isolated from patients, might create new opportunities for the generation of more specific and personalised therapies. However, these systems are unable to recapitulate the tumour microenvironment and interactions with the immune system, both proven to be critical influences on therapeutic outcomes. Patient-derived xenografts, in particular with humanised mouse models, more faithfully mimic the physiology of human liver cancer but are costly and time-consuming, which can be prohibitive for personalising therapies in the setting of an aggressive malignancy. In this review, we discuss the latest advances in the development of more accurate preclinical models to better understand liver cancer biology and identify paradigm-changing therapies, stressing the importance of a bi-directional communicative flow between clinicians and researchers to establish reliable model systems and determine how best to apply them to expanding our current knowledge.
Collapse
|
106
|
Lu Y, Min Z, Qin A, Wu J, Jiang X, Qiao Z. Role of miR-18a and miR-25 disruption and its mechanistic pattern in progression of liver cancer. 3 Biotech 2020; 10:74. [PMID: 32051807 DOI: 10.1007/s13205-020-2064-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 01/08/2020] [Indexed: 12/14/2022] Open
Abstract
This study examined the molecular mechanisms underlying the roles of the microRNAs miR-18a and miR-25 in the progression of human liver cancer. Liver cancer biopsies obtained from early-stage liver cancer patients were examined by qRT-PCR and Northern blotting to examine the expression of miR-18a and miR-25. Both microRNAs were overexpressed in mouse primary hepatocytes following transfection of the cells with vectors encoding the microRNAs. An analysis of biopsy samples from liver cancer patients indicated that both miR-18a and miR-25 were overexpressed during the early stages of liver cancer. Further, qRT-PCR and Northern blotting confirmed that both of these microRNAs play crucial roles in the progression of liver cancer. Our findings clearly indicate that miR-18a and miR-25 can be used as prognostic biomarkers for early-stage liver cancer. Hence, miR-18a and miR-25 may have value as prognostic indicators and may facilitate the development of novel therapeutics for liver cancer.
Collapse
Affiliation(s)
- Yijie Lu
- Department of General Surgery, Nanjing Medical University, Affiliated Suzhou Hospital, Suzhou Municipal Hospital, 26 Daoqian Street, Gusu, Suzhou, 215000 Jiangsu People's Republic of China
| | - Zhai Min
- Department of General Surgery, Nanjing Medical University, Affiliated Suzhou Hospital, Suzhou Municipal Hospital, 26 Daoqian Street, Gusu, Suzhou, 215000 Jiangsu People's Republic of China
| | - Ancheng Qin
- Department of General Surgery, Nanjing Medical University, Affiliated Suzhou Hospital, Suzhou Municipal Hospital, 26 Daoqian Street, Gusu, Suzhou, 215000 Jiangsu People's Republic of China
| | - Jianwu Wu
- Department of General Surgery, Nanjing Medical University, Affiliated Suzhou Hospital, Suzhou Municipal Hospital, 26 Daoqian Street, Gusu, Suzhou, 215000 Jiangsu People's Republic of China
| | - Xinwei Jiang
- Department of General Surgery, Nanjing Medical University, Affiliated Suzhou Hospital, Suzhou Municipal Hospital, 26 Daoqian Street, Gusu, Suzhou, 215000 Jiangsu People's Republic of China
| | - Zhiming Qiao
- Department of General Surgery, Nanjing Medical University, Affiliated Suzhou Hospital, Suzhou Municipal Hospital, 26 Daoqian Street, Gusu, Suzhou, 215000 Jiangsu People's Republic of China
| |
Collapse
|
107
|
Whitlock RS, Yang T, Vasudevan SA, Woodfield SE. Animal Modeling of Pediatric Liver Cancer. Cancers (Basel) 2020; 12:cancers12020273. [PMID: 31979130 PMCID: PMC7072332 DOI: 10.3390/cancers12020273] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/19/2020] [Accepted: 01/19/2020] [Indexed: 01/09/2023] Open
Abstract
Hepatoblastoma (HB) is the most common pediatric liver malignancy. Management of HB requires multidisciplinary efforts. The 5-year overall survival of this disease is about 80% in developed countries. Despite advances in the care of these patients, survival in recurrent or treatment-refractory disease is lower than 50%. This is due to more complex tumor biology, including hepatocellular carcinoma (HCC)-like mutations and expression of aggressive gene signatures leading to chemoresistance, vascular invasion, and metastatic spread. The current treatment protocols for pediatric liver cancer do not incorporate targeted therapies, and the ability to test these therapies is limited due to the inaccessibility of cell lines and mouse models. In this review, we discuss the current status of preclinical animal modeling in pediatric liver cancer, primarily HB. Although HB is a rare cancer, the research community has worked together to develop a range of interesting and relevant mouse models for diverse preclinical studies.
Collapse
Affiliation(s)
- Richard S. Whitlock
- Divisions of Pediatric Surgery and Surgical Research, Michael E. DeBakey Department of Surgery, Pediatric Surgical Oncology Laboratory, Texas Children’s Surgical Oncology Program, Texas Children’s Liver Tumor Program, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; (R.S.W.); (S.A.V.)
| | - Tianyou Yang
- Department of Pediatric Surgery, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Sanjeev A. Vasudevan
- Divisions of Pediatric Surgery and Surgical Research, Michael E. DeBakey Department of Surgery, Pediatric Surgical Oncology Laboratory, Texas Children’s Surgical Oncology Program, Texas Children’s Liver Tumor Program, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; (R.S.W.); (S.A.V.)
| | - Sarah E. Woodfield
- Divisions of Pediatric Surgery and Surgical Research, Michael E. DeBakey Department of Surgery, Pediatric Surgical Oncology Laboratory, Texas Children’s Surgical Oncology Program, Texas Children’s Liver Tumor Program, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; (R.S.W.); (S.A.V.)
- Correspondence: ; Tel.: +1-832-824-4591
| |
Collapse
|
108
|
Overexpression of Hepatocyte Chemerin-156 Lowers Tumor Burden in a Murine Model of Diethylnitrosamine-Induced Hepatocellular Carcinoma. Int J Mol Sci 2019; 21:ijms21010252. [PMID: 31905933 PMCID: PMC6982125 DOI: 10.3390/ijms21010252] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 12/20/2019] [Accepted: 12/26/2019] [Indexed: 12/13/2022] Open
Abstract
The tumor inhibitory potential of the highly active chemerin-156 isoform was described in orthotopic models of hepatocellular carcinoma (HCC). The majority of HCC arises in the fibrotic liver, which was not reproduced in these studies. Here, a potential therapeutic activity of chemerin-156 was evaluated in diethylnitrosamine (DEN)-induced liver cancer, which mimics fibrosis-associated HCC. Mice were infected with adeno-associated virus (AAV) six months after DEN injection to overexpress chemerin-156 in the liver, and animals injected with non-recombinant-AAV served as controls. Three months later, the animals were killed. Both groups were comparable with regard to liver steatosis and fibrosis. Of note, the number of very small tumors was reduced by chemerin-156. Anyhow, the expression of inflammatory and profibrotic genes was similar in larger tumors of control and chemerin-156-AAV-infected animals. Although genes with a role in lipid metabolism, like 3-hydroxy-3-methylglutaryl-coenzym-A--reductase, were overexpressed in tumors of animals with high chemerin-156, total hepatic cholesterol, diacylglycerol and triglyceride levels, and distribution of individual lipid species were normal. Chemerin-156-AAV-infected mice had elevated hepatic and systemic chemerin. Ex vivo activation of the chemerin receptor chemokine-like receptor 1 increased in parallel with serum chemerin, illustrating the biological activity of the recombinant protein. In the tumors, chemerin-155 was the most abundant variant. Chemerin-156 was not detected in tumors of the controls and was hardly found in chemerin-156-AAV infected animals. In conclusion, the present study showed that chemerin-156 overexpression caused a decline in the number of small lesions but did not prevent the growth of pre-existing neoplasms.
Collapse
|
109
|
Oroxin B Induces Apoptosis by Down-Regulating MicroRNA-221 Resulting in the Inactivation of the PTEN/PI3K/AKT Pathway in Liver Cancer. Molecules 2019; 24:molecules24234384. [PMID: 31801250 PMCID: PMC6930563 DOI: 10.3390/molecules24234384] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 11/25/2019] [Accepted: 11/28/2019] [Indexed: 02/07/2023] Open
Abstract
This study aims to investigate the anticancer effect of Oroxin B (OB) both in vitro and in vivo, and the molecular mechanism involved in microRNA-221 and the PI3K/Akt/PTEN pathway through modulation of apoptosis in Hepatocellular carcinoma (HCC). DEN-induced rats and HepG2 cells based on the microfluidic chip were employed, while the mRNA and protein expression of microRNA-221, PI3K, p-Akt and PTEN were evaluated by RT-PCR and Western blot analysis. Based on Microfluidic Chip and DEN-induced rat model, OB effectively exerts anti-liver cancer effect both in vitro and in vivo, and the expression of miR-221 in OB treated groups was significantly lower than that in the control group (** p < 0.01). The RT-PCR and Western blot results suggested the PI3K mRNA and protein in OB treated groups were both lower than those in control group and indicated the overexpression of PTEN. Therefore, OB effectively exerts anticancer effects by positively regulating the PTEN gene and then inactivating the PI3K/Akt signaling pathway through down-regulating the expression of the microRNA-221, thereby inducing apoptosis of liver cancer cells. This study offers a theoretical evidence for further development and clinical guidance of OB as an anti-tumor agent.
Collapse
|
110
|
HMGCS2 Mediates Ketone Production and Regulates the Proliferation and Metastasis of Hepatocellular Carcinoma. Cancers (Basel) 2019; 11:cancers11121876. [PMID: 31779269 PMCID: PMC6966636 DOI: 10.3390/cancers11121876] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 11/22/2019] [Accepted: 11/22/2019] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary malignant tumor worldwide; however, the traditional therapeutic approaches and survival rates are still limited. To improve current therapies, it is necessary to investigate the molecular mechanisms underlying liver cancer and to identify potential therapeutic targets. The aims of this study were to verify the mechanisms and therapeutic potential of the ketogenesis rate-limiting enzyme 3-Hydroxymethylglutaryl-CoA synthase 2 (HMGCS2) in HCC. Immunohistochemical staining of human liver disease tissue arrays showed that HMGCS2 is abundantly expressed in normal liver tissues but is downregulated in cirrhosis and HCC tissues. In HCC patients, lower HMGCS2 expression was correlated with higher pathological grades and clinical stages. In our investigation of the molecular mechanisms of HMGCS2 in HCC, we showed that knockdown of HMGCS2 decreased ketone production, which promoted cell proliferation, cell migration, and xenograft tumorigenesis by enhancing c-Myc/cyclinD1 and EMT signaling and by suppressing the caspase-dependent apoptosis pathway. Ketone body treatment reduced the proliferation- and migration-promoting effects of HMGCS2 knockdown in cells. In contrast, HMGCS2 overexpression increased the intracellular ketone level and inhibited cell proliferation, cell migration, and xenograft tumorigenesis. Finally, ketogenic diet administration significantly inhibited liver cancer cell growth in mice. Our studies highlight the potential therapeutic strategy of targeting HMGCS2-mediated ketogenesis in liver cancer.
Collapse
|
111
|
Mo'men YS, Hussein RM, Kandeil MA. A novel chemoprotective effect of tiopronin against diethylnitrosamine-induced hepatocellular carcinoma in rats: Role of ASK1/P38 MAPK-P53 signalling cascade. Clin Exp Pharmacol Physiol 2019; 47:322-332. [PMID: 31663622 DOI: 10.1111/1440-1681.13204] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 10/18/2019] [Accepted: 10/28/2019] [Indexed: 12/16/2022]
Abstract
Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related death worldwide. Oxidative stress contributes significantly to HCC pathogenesis. In this study, we investigated the possible chemoprotective effect of the thiol group-containing compound, tiopronin, against HCC induced chemically by diethylnitrosamine (DENA) in rats. In addition, we elucidated the possible underlying molecular mechanism. Adult male Wistar rats were divided into: Control group, DENA-treated group and tiopronin + DENA-treated group. Liver function tests (ALT, AST, ALP, albumin, total and direct bilirubin) as well as alpha fetoprotein (AFP) concentration were measured in the sera of samples. Oxidative stress biomarkers such as malondialdehyde, nitric oxide, catalase and glutathione peroxidase were measured in the liver tissue homogenates. Determination of the phosphorylated apoptosis signal-regulating kinase 1 (phospho-ASK1), phospho-P38 and phospho-P53 proteins by western blotting, caspase 3 by immunofluorescence in addition to histopathological examination of the liver tissues were performed. Our results showed that tiopronin prevented the DENA-induced elevation of the liver function enzymes and AFP. It also preserved the activities of antioxidant enzymes as well as providing protection from the appearance of HCC histopathological features. Interestingly, tiopronin significantly decreased the expression level of phospho-ASK1, phospho-P38 and phospho-P53, caspase 3 in the liver tissues. These novel findings suggested that tiopronin is an antioxidant drug with a chemoprotective effect against DENA-induced HCC through maintaining the normal activity of ASK1/ P38 MAPK/ P53 signalling pathway.
Collapse
Affiliation(s)
- Yomna S Mo'men
- Department of Biochemistry, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Rasha M Hussein
- Department of Biochemistry, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt.,Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, Mutah University, Al-Karak, Jordan
| | - Mohamed A Kandeil
- Department of Biochemistry, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, Egypt
| |
Collapse
|
112
|
Mohamed DI, Khairy E, Khedr SA, Habib EK, Elayat WM, El-Kharashi OA. N-acetylcysteine (NAC) alleviates the peripheral neuropathy associated with liver cirrhosis via modulation of neural MEG3/PAR2/ NF-ҡB axis. Neurochem Int 2019; 132:104602. [PMID: 31751619 DOI: 10.1016/j.neuint.2019.104602] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 11/07/2019] [Accepted: 11/11/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIM Oxidative stress (OS) is accused in pathogenesis of many diseases, including liver cirrhosis by many mechanisms. One of them is the disturbance of long non coding maternally expressed 3 (MEG3)/protease activated receptor 2 (PAR2) downstream pathway. We aimed to investigate the role of this axis in cirrhotic neuropathy and whether an antioxidant compound such as N-acetylcysteine (NAC) could improve the peripheral nerve function through repression of MEG3/PAR2. METHODS Thirty Wistar rats were used and divided into 5 groups; naive, thiacetamide (TAA) (200 mg/kg 3 times/week. i.p. for 8 weeks) and TAA+NAC (50 or 100 or 200 mg/kg/day) groups. Von Frey (VF) test for mechanical nociceptive responses, hepatic& neural MEG3, NF-ҡB and neural PAR2 expression by PCR, histological studies for liver and sciatic nerve together with the dorsopedal skin thickness were done. RESULTS TAA induced significant decrease in liver function, negative VF test, an increase in the expression of hepatic& neural MEG3, NF-ҡB and neural PAR2. The histological studies showed cirrhotic changes with atrophy of the sciatic nerve and the dorsal skin. NAC improved the liver function together with reversal of the neural: functional, biochemical and histological changes in a dose dependent manner. CONCLUSIONS NAC could improve the peripheral neuropathy in cirrhotic rat through suppression of MEG3/PAR2 expression.
Collapse
Affiliation(s)
- Doaa I Mohamed
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| | - Eman Khairy
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| | - Sara A Khedr
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| | - Eman K Habib
- Department of Anatomy and Embryology, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| | - Wael M Elayat
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| | - Omnyah A El-Kharashi
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
113
|
Mouse Models for Immunotherapy in Hepatocellular Carcinoma. Cancers (Basel) 2019; 11:cancers11111800. [PMID: 31731753 PMCID: PMC6896030 DOI: 10.3390/cancers11111800] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 11/01/2019] [Indexed: 12/13/2022] Open
Abstract
Liver cancer is one of the dominant causes of cancer-related mortality, and the survival rate of liver cancer is among the lowest for all cancers. Immunotherapy for hepatocellular carcinoma (HCC) has yielded some encouraging results, but the percentage of patients responding to single-agent therapies remains low. Therefore, potential directions for improved immunotherapies include identifying new immune targets and checkpoints and customizing treatment procedures for individual patients. The development of combination therapies for HCC is also crucial and urgent and, thus, further studies are required. Mice have been utilized in immunotherapy research due to several advantages, for example, being low in cost, having high success rates for inducing tumor growth, and so on. Moreover, immune-competent mice are used in immunotherapy research to clarify the role that the immune system plays in cancer growth. In this review paper, the advantages and disadvantages of mouse models for immunotherapy, the equipment that are used for monitoring HCC, and the cell strains used for inducing HCC are reviewed.
Collapse
|
114
|
Animal Models of Hepatocellular Carcinoma Prevention. Cancers (Basel) 2019; 11:cancers11111792. [PMID: 31739536 PMCID: PMC6895981 DOI: 10.3390/cancers11111792] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 11/08/2019] [Accepted: 11/10/2019] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a deadly disease and therapeutic efficacy in advanced HCC is limited. Since progression of chronic liver disease to HCC involves a long latency period of a few decades, a significant window of therapeutic opportunities exists for prevention of HCC and improve patient prognosis. Nonetheless, there has been no clinical advancement in instituting HCC chemopreventive strategies. Some of the major challenges are heterogenous genetic aberrations of HCC, significant modulation of tumor microenvironment and incomplete understanding of HCC tumorigenesis. To this end, animal models of HCC are valuable tools to evaluate biology of tumor initiation and progression with specific insight into molecular and genetic mechanisms involved. In this review, we describe various animal models of HCC that facilitate effective ways to study therapeutic prevention strategies that have translational potential to be evaluated in a clinical context.
Collapse
|
115
|
Genomic Perspective on Mouse Liver Cancer Models. Cancers (Basel) 2019; 11:cancers11111648. [PMID: 31731480 PMCID: PMC6895968 DOI: 10.3390/cancers11111648] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 10/22/2019] [Accepted: 10/23/2019] [Indexed: 02/07/2023] Open
Abstract
Selecting the most appropriate mouse model that best recapitulates human hepatocellular carcinoma (HCC) allows translation of preclinical mouse studies into clinical studies. In the era of cancer genomics, comprehensive and integrative analysis of the human HCC genome has allowed categorization of HCC according to molecular subtypes. Despite the variety of mouse models that are available for preclinical research, there is a lack of evidence for mouse models that closely resemble human HCC. Therefore, it is necessary to identify the accurate mouse models that represent human HCC based on molecular subtype as well as histologic aggressiveness. In this review, we summarize the mouse models integrated with human HCC genomic data to provide information regarding the models that recapitulates the distinct aspect of HCC biology and prognosis based on molecular subtypes.
Collapse
|
116
|
Dey P, Kim JB, Chitchumroonchokchai C, Li J, Sasaki GY, Olmstead BD, Stock KL, Thomas-Ahner JM, Clinton SK, Bruno RS. Green tea extract inhibits early oncogenic responses in mice with nonalcoholic steatohepatitis. Food Funct 2019; 10:6351-6361. [PMID: 31503268 DOI: 10.1039/c9fo01199d] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nonalcoholic steatohepatitis (NASH) increases hepatocellular carcinoma (HCC) risk. We hypothesized that the hepatoprotective anti-inflammatory benefits of catechin-rich green tea extract (GTE) would protect against HCC progression by inhibiting NASH-associated liver injury and pro-oncogenic responses. We used an HCC model in high-fat (HF)-fed mice that mimics early oncogenic events during NASH without inducing tumorigenesis and premature mortality. Male C57BL/6J mice (4-weeks old) were fed a HF diet containing GTE at 0% or 2%. Mice were administered saline or diethylnitrosamine (DEN; 60 mg kg-1, i.p.) at 5-weeks and 7-weeks of age. NASH, inflammation, fibrosis, and oncogenic responses were assessed at 25-weeks of age. Saline-treated mice showed prominent histopathological signs of steatosis and hepatocellular ballooning. Although DEN did not impact adiposity, steatosis, ballooning and hepatic lipid accumulation, these parameters were attenuated by GTE regardless of DEN. Hepatic lipid peroxidation and fibrosis that were increased by DEN were attenuated by GTE. Hepatic TLR4, MCP1 and TNFα mRNA levels were unaffected by DEN, whereas iNOS was increased by DEN. These transcripts were lowered by GTE. GTE attenuated the frequency of PCNA+ hepatocytes and mRNA expression of cyclin D1, MIB1 and Ki-67 that were otherwise increased by DEN. GTE increase APAF1 mRNA that was otherwise lowered by DEN. Relative to saline-treated mice, DEN increased mRNA levels of oncostatin M, gp130, c-Fos, c-Myc and survivin; each was lowered by GTE in DEN-treated mice. These findings indicate that GTE may protect against hepatic oncogenesis by limiting early steps in the carcinogenic cascade related to NASH-associated HCC.
Collapse
Affiliation(s)
- Priyankar Dey
- Human Nutrition Program, The Ohio State University, Columbus, OH, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
117
|
Mancarella S, Krol S, Crovace A, Leporatti S, Dituri F, Frusciante M, Giannelli G. Validation of Hepatocellular Carcinoma Experimental Models for TGF-β Promoting Tumor Progression. Cancers (Basel) 2019; 11:E1510. [PMID: 31600917 PMCID: PMC6826694 DOI: 10.3390/cancers11101510] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 09/27/2019] [Accepted: 09/29/2019] [Indexed: 02/07/2023] Open
Abstract
Transforming growth factor beta (TGF-β) is a pleiotropic cytokine with dual role in hepatocellular carcinoma (HCC). It acts as tumor-suppressor and tumor-promoter in the early and late stage respectively. TGF-β influences the tumor-stroma cross-talk affecting the tumoral microenvironment. Therefore, inhibiting the TGF- β mediated pathway alone and/or in combination with chemotherapeutics represents an important therapeutic option. Experimental models to dissect the role of TGF-β in HCC tumor progression as well as the effectiveness of specific inhibitors are tricky. HCC cell lines respond to TGF-β according to their epithelial phenotype. However, the mesenchymal and more aggressive HCC cell lines in vitro, do not develop tumors when transplanted in vivo, thus hampering the understanding of molecular pathways that dictate outcome. In addition, in this model the native immune system is abolished, therefore the contribution of inflammation in hepatocarcinogenesis is unreliable. Different strategies have been set up to engineer HCC animal models, including genetically modified mice, chemically induced HCC, or hydrodynamic techniques. Patient-derived xenograft is currently probably the most fascinating model, keeping in mind that models cannot mirror all the reality. In this context, we discuss the different available HCC mouse models including our experimental model treated with inhibitor of TGF-β receptor Type I kinase (Galunisertib) and a potential role of exosomes in TGF-β moderated tumor progression of HCC. Unfortunately, no positive results were obtained in our treated orthotopic model because it does not reproduce the critical tumor-stroma interactions of the HCC.
Collapse
Affiliation(s)
- Serena Mancarella
- National Institute of Gastroenterology, "S. de Bellis" Research Hospital, Castellana Grotte, Bari 70013, Italy.
| | - Silke Krol
- National Institute of Gastroenterology, "S. de Bellis" Research Hospital, Castellana Grotte, Bari 70013, Italy.
| | - Alberto Crovace
- National Institute of Gastroenterology, "S. de Bellis" Research Hospital, Castellana Grotte, Bari 70013, Italy.
| | | | - Francesco Dituri
- National Institute of Gastroenterology, "S. de Bellis" Research Hospital, Castellana Grotte, Bari 70013, Italy.
| | - Martina Frusciante
- National Institute of Gastroenterology, "S. de Bellis" Research Hospital, Castellana Grotte, Bari 70013, Italy.
| | - Gianluigi Giannelli
- National Institute of Gastroenterology, "S. de Bellis" Research Hospital, Castellana Grotte, Bari 70013, Italy.
| |
Collapse
|
118
|
Martin-Levilain J, Li L, Maechler P. Reply to Mishra: Prohibitin heterodimers-a complex time dependence for carcinogenesis. J Biol Chem 2019; 294:14837. [PMID: 31586027 DOI: 10.1074/jbc.rl119.010819] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- Juliette Martin-Levilain
- Department of Cell Physiology and Metabolism, University of Geneva Medical Centre, 1206 Geneva, Switzerland.,Faculty Diabetes Centre, University of Geneva Medical Centre, 1206 Geneva, Switzerland
| | - Lingzi Li
- Department of Cell Physiology and Metabolism, University of Geneva Medical Centre, 1206 Geneva, Switzerland.,Faculty Diabetes Centre, University of Geneva Medical Centre, 1206 Geneva, Switzerland
| | - Pierre Maechler
- Department of Cell Physiology and Metabolism, University of Geneva Medical Centre, 1206 Geneva, Switzerland .,Faculty Diabetes Centre, University of Geneva Medical Centre, 1206 Geneva, Switzerland
| |
Collapse
|
119
|
Animal Models of Hepatocellular Carcinoma: The Role of Immune System and Tumor Microenvironment. Cancers (Basel) 2019; 11:cancers11101487. [PMID: 31581753 PMCID: PMC6826986 DOI: 10.3390/cancers11101487] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 09/28/2019] [Accepted: 09/30/2019] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common type of liver cancer in adults and has one of the highest mortality rates of solid cancers. Ninety percent of HCCs are associated with liver fibrosis or cirrhosis developed from chronic liver injuries. The immune system of the liver contributes to the severity of the necrotic-inflammatory tissue damage, the establishment of fibrosis and cirrhosis, and the disease progression towards HCC. Immunotherapies have emerged as an exciting strategy for HCC treatment, but their effect is limited, and an extensive translation research is urgently needed to enhance anti-tumor efficacy and clinical success. Establishing HCC animal models that are analogous to human disease settings, i.e., mimicking the tumor microenvironment of HCC, is extremely challenging. Hence, this review discusses different animal models of HCC by summarizing their advantages and their limits with a specific focus on the role of the immune system and tumor microenvironment.
Collapse
|
120
|
Vitobello A, Perner J, Beil J, Zhu J, Del Río-Espínola A, Morawiec L, Westphal M, Dubost V, Altorfer M, Naumann U, Mueller A, Kapur K, Borowsky M, Henderson C, Wolf CR, Schwarz M, Moggs J, Terranova R. Drug-induced chromatin accessibility changes associate with sensitivity to liver tumor promotion. Life Sci Alliance 2019; 2:e201900461. [PMID: 31615920 PMCID: PMC6795216 DOI: 10.26508/lsa.201900461] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 09/26/2019] [Accepted: 09/26/2019] [Indexed: 12/27/2022] Open
Abstract
Liver cancer susceptibility varies amongst humans and between experimental animal models because of multiple genetic and epigenetic factors. The molecular characterization of such susceptibilities has the potential to enhance cancer risk assessment of xenobiotic exposures and disease prevention strategies. Here, using DNase I hypersensitivity mapping coupled with transcriptomic profiling, we investigate perturbations in cis-acting gene regulatory elements associated with the early stages of phenobarbital (PB)-mediated liver tumor promotion in susceptible versus resistant mouse strains (B6C3F1 versus C57BL/6J). Integrated computational analyses of strain-selective changes in liver chromatin accessibility underlying PB response reveal differential epigenetic regulation of molecular pathways associated with PB-mediated tumor promotion, including Wnt/β-catenin signaling. Complementary transcription factor motif analyses reveal mouse strain-selective gene regulatory networks and a novel role for Stat, Smad, and Fox transcription factors in the early stages of PB-mediated tumor promotion. Mapping perturbations in cis-acting gene regulatory elements provides novel insights into the molecular basis for susceptibility to xenobiotic-induced rodent liver tumor promotion and has the potential to enhance mechanism-based cancer risk assessments of xenobiotic exposures.
Collapse
Affiliation(s)
- Antonio Vitobello
- Novartis Institutes for BioMedical Research (NIBR), Basel, Switzerland
- Inserm, Unité Mixte de Recherche (UMR) 1231, Université de Bourgogne-Franche Comté, Dijon, France
| | - Juliane Perner
- Novartis Institutes for BioMedical Research (NIBR), Basel, Switzerland
| | - Johanna Beil
- Novartis Institutes for BioMedical Research (NIBR), Basel, Switzerland
| | | | | | - Laurent Morawiec
- Novartis Institutes for BioMedical Research (NIBR), Basel, Switzerland
| | | | - Valérie Dubost
- Novartis Institutes for BioMedical Research (NIBR), Basel, Switzerland
| | - Marc Altorfer
- Novartis Institutes for BioMedical Research (NIBR), Basel, Switzerland
| | - Ulrike Naumann
- Novartis Institutes for BioMedical Research (NIBR), Basel, Switzerland
| | - Arne Mueller
- Novartis Institutes for BioMedical Research (NIBR), Basel, Switzerland
| | - Karen Kapur
- Novartis Institutes for BioMedical Research (NIBR), Basel, Switzerland
| | | | - Colin Henderson
- School of Medicine, Jacqui Wood Cancer Centre, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
- Innovative Medicines Initiative MARCAR Consortium (http://www.imi-marcar.eu/index.php)
| | - C Roland Wolf
- School of Medicine, Jacqui Wood Cancer Centre, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
- Innovative Medicines Initiative MARCAR Consortium (http://www.imi-marcar.eu/index.php)
| | - Michael Schwarz
- Department of Toxicology, University of Tübingen, Tübingen, Germany
- Innovative Medicines Initiative MARCAR Consortium (http://www.imi-marcar.eu/index.php)
| | - Jonathan Moggs
- Novartis Institutes for BioMedical Research (NIBR), Basel, Switzerland
- Innovative Medicines Initiative MARCAR Consortium (http://www.imi-marcar.eu/index.php)
| | - Rémi Terranova
- Novartis Institutes for BioMedical Research (NIBR), Basel, Switzerland
| |
Collapse
|
121
|
Zhou ZF, Peng F, Li JY, Ye YB. Intratumoral IL-12 Gene Therapy Inhibits Tumor Growth In A HCC-Hu-PBL-NOD/SCID Murine Model. Onco Targets Ther 2019; 12:7773-7784. [PMID: 31571927 PMCID: PMC6760038 DOI: 10.2147/ott.s222097] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 09/11/2019] [Indexed: 12/18/2022] Open
Abstract
Purpose This study aimed to evaluate the efficacy and safety of intratumoral IL-12 gene therapy in an HCC-hu-PBL-NOD/SCID mouse model. Materials and methods The HCC murine model was generated in NOD/SCID mice, and mice with grafted tumors were injected intraperitoneally with 2 × 107 human peripheral blood lymphocytes 14 days after modeling. After 4 days, mice were randomly divided into the 9597/IL-12 group, the 9597/plasmid group and the PBS group. The changes of tumor volume were measured and mouse peripheral blood was sampled post-treatment for ELISA and CBA analyses, and the grafted tumors were collected 28 days post-treatment for immunohistochemistry, ELISA, CBA and detection of cell cycle and apoptosis. Results The tumor volume was smaller in the 9597/IL-12 group than in the 9597/plasmid and PBS groups on days 7, 14, 21, and 28 post-treatment (P < 0.05). Higher IL-12 levels were detected in the peripheral blood and the supernatants of grafted tumor homogenates in the 9597/IL-12 group than in the 9597/plasmid and PBS groups 7, 14, 21 and 28 days post-treatment (P < 0.05). IHC revealed higher counts of CD3+T cells, CD4+T helper cells, IFN-γ Th1 cells+ and S-100 protein positive dentric cells and lower MVD in the 9597/IL-12 group than in the 9597/plasmid and PBS groups (P < 0.05). Flow cytometry showed a significantly higher proportion of HCC cells at the G0/G1 phase and a significantly lower proportion of HCC cells at the S phase in the 9597/IL-12 group than in the PBS group (P < 0.05) and a greater apoptotic rate of HCC cells in the 9597/IL-12 group than in the 9597/plasmid and PBS groups (P < 0.05). Conclusion Intratumoral IL-12 gene therapy may inhibit tumorigenesis with mild adverse effects in a HCC-hu-PBL-NOD/SCID murine model through inhibiting angiogenesis, arresting cells in G0/G1 phase and inducing apoptosis.
Collapse
Affiliation(s)
- Zhi-Feng Zhou
- Laboratory of Immuno-Oncology, Fujian Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou 350014, People's Republic of China.,Fujian Key Laboratory of Translational Cancer Medicine, Fujian Cancer Hospital, Fuzhou 350014, People's Republic of China
| | - Feng Peng
- Laboratory of Immuno-Oncology, Fujian Medical University Cancer Hospital, Fuzhou 350014, People's Republic of China
| | - Jie-Yu Li
- Laboratory of Immuno-Oncology, Fujian Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou 350014, People's Republic of China.,Fujian Key Laboratory of Translational Cancer Medicine, Fujian Cancer Hospital, Fuzhou 350014, People's Republic of China
| | - Yun-Bin Ye
- Laboratory of Immuno-Oncology, Fujian Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou 350014, People's Republic of China.,Fujian Key Laboratory of Translational Cancer Medicine, Fujian Cancer Hospital, Fuzhou 350014, People's Republic of China
| |
Collapse
|
122
|
Qi X, Schepers E, Avella D, Kimchi ET, Kaifi JT, Staveley-O'Carroll KF, Li G. An Oncogenic Hepatocyte-Induced Orthotopic Mouse Model of Hepatocellular Cancer Arising in the Setting of Hepatic Inflammation and Fibrosis. J Vis Exp 2019. [PMID: 31566616 DOI: 10.3791/59368] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The absence of a clinically relevant animal model addressing the typical immune characteristics of hepatocellular cancer (HCC) has significantly impeded elucidation of the underlying mechanisms and development of innovative immunotherapeutic strategies. To develop an ideal animal model recapitulating human HCC, immunocompetent male C57BL/6J mice first receive a carbon tetrachloride (CCl4) injection to induce liver fibrosis, then receive histologically-normal oncogenic hepatocytes from young male SV40 T antigen (TAg)-transgenic mice (MTD2) by intra-splenic (ISPL) inoculation. Androgen generated in recipient male mice at puberty initiates TAg expression under control of a liver-specific promoter. As a result, the transferred hepatocytes become cancer cells and form tumor masses in the setting of liver fibrosis/cirrhosis. This novel model mimics human HCC initiation and progression in the context of liver fibrosis/cirrhosis and reflects the most typical features of human HCC including immune dysfunction.
Collapse
Affiliation(s)
- Xiaoqiang Qi
- Department of Surgery, University of Missouri-Columbia; Ellis Fischel Cancer Center, University of Missouri-Columbia; Molecular Microbiology and Immunology, University of Missouri-Columbia
| | - Emily Schepers
- Department of Surgery, University of Missouri-Columbia; Ellis Fischel Cancer Center, University of Missouri-Columbia
| | - Diego Avella
- Department of Surgery, University of Missouri-Columbia; Ellis Fischel Cancer Center, University of Missouri-Columbia
| | - Eric T Kimchi
- Department of Surgery, University of Missouri-Columbia; Ellis Fischel Cancer Center, University of Missouri-Columbia
| | - Jussuf T Kaifi
- Department of Surgery, University of Missouri-Columbia; Ellis Fischel Cancer Center, University of Missouri-Columbia
| | - Kevin F Staveley-O'Carroll
- Department of Surgery, University of Missouri-Columbia; Ellis Fischel Cancer Center, University of Missouri-Columbia;
| | - Guangfu Li
- Department of Surgery, University of Missouri-Columbia; Ellis Fischel Cancer Center, University of Missouri-Columbia; Molecular Microbiology and Immunology, University of Missouri-Columbia;
| |
Collapse
|
123
|
Liebig M, Dannenberger D, Vollmar B, Abshagen K. n-3 PUFAs reduce tumor load and improve survival in a NASH-tumor mouse model. Ther Adv Chronic Dis 2019; 10:2040622319872118. [PMID: 31523414 PMCID: PMC6728677 DOI: 10.1177/2040622319872118] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 07/16/2019] [Indexed: 12/22/2022] Open
Abstract
Background With 9.1% of all cancer deaths, hepatocellular carcinoma is the second leading cause of cancer deaths worldwide. Due to the increasing prevalence of metabolic syndrome, nonalcoholic fatty liver disease (NAFLD) has evolved into a major risk factor for hepatocellular carcinoma development. Herein, we investigated whether a dietary n-3 polyunsaturated fatty acid (PUFA) supplementation improves the outcome of progressive NAFLD. Methods Feeding three high-fat diets, differing in n-3 and n-6 PUFA contents and ratios (n-3/n-6: 1:8, 1:1, 5:1), the impact of n-3 PUFAs and n-3/n-6 PUFA ratios on NAFLD-related liver fibrosis and tumorigenesis was analyzed in 12- and 20-week-old streptozotocin/high-fat diet (STZ/HFD)-treated mice. Results Feeding of n-3 PUFA-rich diets (1:1 and 5:1) resulted in increased hepatic n-3 PUFA content and n-3/n-6 PUFA ratio with decreased hepatic lipid accumulation. In 20-week-old mice, n-3 PUFA-rich diets alleviated tumor load significantly, with reduced liver/body weight index, tumor size, and tumor number. Finally, these effects were accompanied by a significant improvement of survival of these mice. Conclusions Herein, we showed that increased n-3 PUFA content and n-3/n-6 PUFA ratios lead to improved survival and attenuated tumor progression in STZ/HFD-treated mice. Thus, n-3 PUFAs could be the basis for new therapeutic options against NAFLD-related tumorigenesis.
Collapse
Affiliation(s)
- Marie Liebig
- Institute for Experimental Surgery, University Medicine Rostock, Germany
| | - Dirk Dannenberger
- Institute of Muscle Biology and Growth, Leibniz Institute for Farm Animal Biology, Dummerstorf, Germany
| | - Brigitte Vollmar
- Institute for Experimental Surgery, University Medicine Rostock, Germany
| | - Kerstin Abshagen
- Institute for Experimental Surgery, University Medicine Rostock, Germany
| |
Collapse
|
124
|
Fuentes-Hernández S, Alarcón-Sánchez BR, Guerrero-Escalera D, Montes-Aparicio AV, Castro-Gil MP, Idelfonso-García OG, Rosas-Madrigal S, Aparicio-Bautista DI, Pérez-Hernández JL, Reyes-Gordillo K, Lakshman MR, Vásquez-Garzón VR, Baltiérrez-Hoyos R, López-González MDL, Sierra-Santoyo A, Villa-Treviño S, Pérez-Carreón JI, Arellanes-Robledo J. Chronic administration of diethylnitrosamine to induce hepatocarcinogenesis and to evaluate its synergistic effect with other hepatotoxins in mice. Toxicol Appl Pharmacol 2019; 378:114611. [DOI: 10.1016/j.taap.2019.114611] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 05/31/2019] [Accepted: 06/03/2019] [Indexed: 02/06/2023]
|
125
|
Li W, Yue F, Dai Y, Shi B, Xu G, Jiang X, Zhou X, Pfeifer GP, Liu L. Suppressor of hepatocellular carcinoma RASSF1A activates autophagy initiation and maturation. Cell Death Differ 2019; 26:1379-1395. [PMID: 30315205 PMCID: PMC6748129 DOI: 10.1038/s41418-018-0211-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 09/17/2018] [Accepted: 09/20/2018] [Indexed: 01/30/2023] Open
Abstract
RASSF1A (Ras association domain family 1 isoform A) is a tumor suppressor and frequently inactivated by promoter hypermethylation in hepatocellular carcinoma (HCC). Autophagy is to degrade misfolded or aggregated proteins and dysfunctional organelles. Autophagy defects enhance oxidative stress and genome instability to promote tumorigenesis. Activating autophagy flux by increasing levels of the RASSF1A-interacting microtubule-associated protein 1 S (MAP1S) leads to suppression of HCC in addition to extending lifespans. Here we tested whether RASSF1A itself functions as a HCC suppressor and activates autophagy similarly as MAP1S does. We show that RASSF1A deletion leads to an acceleration of diethylnitrosamine-induced HCC and a 31% reduction of median survival times in mice. RASSF1A enhances autophagy initiation by suppressing PI3K-AKT-mTOR through the Hippo pathway-regulatory component MST1 and promotes autophagy maturation by recruiting autophagosomes on RASSF1A-stabilized acetylated microtubules through MAP1S. RASSF1A deletion causes a blockade of autophagy flux. Therefore, RASSF1A may suppress HCC and improve survival by activating autophagy flux.
Collapse
Affiliation(s)
- Wenjiao Li
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, 2121 W. Holcombe Blvd., Houston, TX, 77030, USA
| | - Fei Yue
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, 2121 W. Holcombe Blvd., Houston, TX, 77030, USA
| | - Yuan Dai
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, 2121 W. Holcombe Blvd., Houston, TX, 77030, USA
| | - Boyun Shi
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, 2121 W. Holcombe Blvd., Houston, TX, 77030, USA
- The Fifth Affiliated Hospital, Guangzhou Medical University, 510700, Guangzhou, China
| | - Guibin Xu
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, 2121 W. Holcombe Blvd., Houston, TX, 77030, USA
- The Fifth Affiliated Hospital, Guangzhou Medical University, 510700, Guangzhou, China
| | - Xianhan Jiang
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, 2121 W. Holcombe Blvd., Houston, TX, 77030, USA
- The Fifth Affiliated Hospital, Guangzhou Medical University, 510700, Guangzhou, China
| | - Xinke Zhou
- The Fifth Affiliated Hospital, Guangzhou Medical University, 510700, Guangzhou, China.
| | - Gerd P Pfeifer
- Center for Epigenetics, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Leyuan Liu
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, 2121 W. Holcombe Blvd., Houston, TX, 77030, USA.
- The Fifth Affiliated Hospital, Guangzhou Medical University, 510700, Guangzhou, China.
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University, College Station, TX 77843, USA.
| |
Collapse
|
126
|
Zhu H, Ge K, Lu J, Jia C. Growth inhibitor of human hepatic carcinoma HepG2 cells by evodiamine is associated with downregulation of PRAME. Naunyn Schmiedebergs Arch Pharmacol 2019; 392:1551-1560. [DOI: 10.1007/s00210-019-01701-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 07/18/2019] [Indexed: 12/21/2022]
|
127
|
Tanaka H, Horioka K, Yamamoto M, Asari M, Okuda K, Yamazaki K, Shimizu K, Ogawa K. Overproduction of thrombopoietin by BRAFV600E-mutated mouse hepatocytes and contribution of thrombopoietin to hepatocarcinogenesis. Cancer Sci 2019; 110:2748-2759. [PMID: 31301081 PMCID: PMC6726687 DOI: 10.1111/cas.14130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 07/01/2019] [Accepted: 07/10/2019] [Indexed: 01/27/2023] Open
Abstract
In hepatocarcinogenesis induced by diethylnitrosamine (DEN) in B6C3F1 mice, the BrafV637E mutation, corresponding to the human BRAFV600E mutation, plays a pivotal role. The livers of transgenic mice with a hepatocyte‐specific human BRAFV600E mutation weighed 4.5 times more than that of normal mice and consisted entirely of hepatocytes, resembling DEN‐induced preneoplastic hepatocytes. However, these transgenic mice spontaneously died 7 wk after birth, therefore this study aimed to clarify the causes of death. In the transgenic mice, the liver showed thrombopoietin (TPO) overexpression, which is associated with eventual megakaryocytosis and thrombocytosis, and activated platelets were deposited in hepatic sinusoids. TPO was also overexpressed in the DEN‐induced hepatic tumors, and sinusoidal platelet deposition was observed in the hepatic tumors of humans and mice. Podoplanin was expressed in some of the Kupffer cells in the liver of the transgenic mice, indicating that platelet activation occurred via the interaction of podoplanin with C‐type lectin receptor 2 (CLEC‐2) on the platelet membrane. Additionally, erythrocyte dyscrasia and glomerulonephropathy/interstitial pneumonia associated with platelet deposition were observed. In the transgenic mice, aspirin (Asp) administration prevented platelet activation, reduced the liver/body weight ratio, decreased the platelet deposition in the liver, kidney, and lung, and prevented erythrocyte dyscrasia and ameliorated the renal/pulmonary changes. Thrombopoietin overproduction by BRAFV600E‐mutated hepatocytes may contribute to hepatocyte proliferation via thrombocytosis, platelet activation, and the interaction of platelets with hepatic sinusoidal cells, while hematologic, renal, and pulmonary disorders due to aberrant platelet activation may lead to spontaneous death in the transgenic mice.
Collapse
Affiliation(s)
- Hiroki Tanaka
- Department of Legal Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Kie Horioka
- Department of Legal Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Masahiro Yamamoto
- Department of Molecular Cancer Science, School of Medicine, Yamagata University, Yamagata, Japan
| | - Masaru Asari
- Department of Legal Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Katsuhiro Okuda
- Department of Legal Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Kosuke Yamazaki
- Department of Clinical Medicine, Surgery Area, Japanese Red Cross Hokkaido College of Nursing, Kitamai, Japan
| | - Keiko Shimizu
- Department of Legal Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Katsuhiro Ogawa
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| |
Collapse
|
128
|
Hage C, Hoves S, Ashoff M, Schandl V, Hört S, Rieder N, Heichinger C, Berrera M, Ries CH, Kiessling F, Pöschinger T. Characterizing responsive and refractory orthotopic mouse models of hepatocellular carcinoma in cancer immunotherapy. PLoS One 2019; 14:e0219517. [PMID: 31291357 PMCID: PMC6619768 DOI: 10.1371/journal.pone.0219517] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 06/25/2019] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common cancers worldwide and has a high mortality rate due to limited treatment options. Hence, the response of HCC to different cancer immunotherapies is being intensively investigated in clinical trials. Immune checkpoint blockers (ICB) show promising results, albeit for a minority of HCC patients. Mouse models are commonly used to evaluate new therapeutic agents or regimens. However, to make clinical translation more successful, better characterized preclinical models are required. We therefore extensively investigated two immune-competent orthotopic HCC mouse models, namely transplanted Hep-55.1c and transgenic iAST, with respect to morphological, immunological and genetic traits and evaluated both models' responsiveness to immunotherapies. Hep-55.1c tumors were characterized by rich fibrous stroma, high mutational load and pronounced immune cell infiltrates, all of which are features of immune-responsive tumors. These characteristics were less distinct in iAST tumors, though these were highly vascularized. Cell depletion revealed that CD8+ T cells from iAST mice do not affect tumor growth and are tumor tolerant. This corresponds to the failure of single and combined ICB targeting PD-1 and CTLA-4. In contrast, combining anti-PD-1 and anti-CTLA-4 showed significant antitumor efficacy in the Hep-55.1c mouse model. Collectively, our data comprehensively characterize two immune-competent HCC mouse models representing ICB responsive and refractory characteristics. Our characterization confirms these models to be suitable for preclinical investigation of novel cancer immunotherapy approaches that aim to either deepen preexisting immune responses or generate de novo immunity against the tumor.
Collapse
MESH Headings
- Animals
- Antigens, Polyomavirus Transforming/genetics
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Agents, Immunological/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- CD8-Positive T-Lymphocytes/immunology
- CTLA-4 Antigen/antagonists & inhibitors
- CTLA-4 Antigen/immunology
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/immunology
- Cell Line, Tumor/transplantation
- Disease Models, Animal
- Drug Resistance, Neoplasm
- Drug Screening Assays, Antitumor/methods
- Female
- Humans
- Liver Neoplasms/drug therapy
- Liver Neoplasms/genetics
- Liver Neoplasms/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Programmed Cell Death 1 Receptor/immunology
- Treatment Outcome
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Carina Hage
- Roche Innovation Center Munich, Roche Pharmaceutical Research and Early Development, Penzberg, Germany
- Institute for Experimental Molecular Imaging, University Clinic and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Sabine Hoves
- Roche Innovation Center Munich, Roche Pharmaceutical Research and Early Development, Penzberg, Germany
| | - Mailin Ashoff
- Roche Innovation Center Munich, Roche Pharmaceutical Research and Early Development, Penzberg, Germany
| | - Veronika Schandl
- Roche Innovation Center Munich, Roche Pharmaceutical Research and Early Development, Penzberg, Germany
| | - Stefan Hört
- Roche Innovation Center Munich, Roche Pharmaceutical Research and Early Development, Penzberg, Germany
| | - Natascha Rieder
- Roche Innovation Center Munich, Roche Pharmaceutical Research and Early Development, Penzberg, Germany
| | - Christian Heichinger
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| | - Marco Berrera
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| | - Carola H. Ries
- Roche Innovation Center Munich, Roche Pharmaceutical Research and Early Development, Penzberg, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, University Clinic and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Thomas Pöschinger
- Roche Innovation Center Munich, Roche Pharmaceutical Research and Early Development, Penzberg, Germany
| |
Collapse
|
129
|
Su XY, Zhao JQ, Li N, Kumar M, yang AMO. Chemoprotective Effects of Resveratrol Against Diethylnitrosamine Induced Hepatocellular Carcinoma in Wistar Rats. INT J PHARMACOL 2019. [DOI: 10.3923/ijp.2019.549.559] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
130
|
Bou About G, Thiebault E, Wattenhofer-Donzé M, Jacobs H, Guimond A, Sorg T, Robinet E, Baumert TF, Monassier L, Herault Y. Ultrasound-Guided Approaches to Improve Orthotopic Mouse Xenograft Models for Hepatocellular Carcinoma. ACTA ACUST UNITED AC 2019; 9:e62. [PMID: 31145554 PMCID: PMC7613417 DOI: 10.1002/cpmo.62] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Hepatocellular carcinoma (HCC) is the second leading cause of cancer death worldwide. While curative approaches for early stage HCC exist, effective treatment options for advanced HCC are lacking. Furthermore, there are no efficient chemopreventive strategies to limit HCC development once cirrhosis is established. One challenge for drug development is unsatisfactory animal models. In this article, we describe an orthotopic xenograft mouse model of human liver cancer cell lines through image-guided injection into the liver. This technique provides a less invasive yet highly efficient approach to engraft human HCC into mouse liver. Similarly, image-guided injections are used to deliver chemotherapeutics locally, enabling reduction in potential systemic adverse effects, while reducing the required dose for a therapeutic effect. In summary, this image-guided strategy provides a novel and convenient approach to improve current HCC mouse models. © 2019 The Authors. This is an open access article under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License, which permits use and distribution in any medium, provided the original work is properly cited, the use is non-commercial and no modifications or adaptations are made.
Collapse
Affiliation(s)
- Ghina Bou About
- Institut Clinique de la Souris, PHENOMIN-ICS, Illkirch, France
| | | | | | - Hugues Jacobs
- Institut Clinique de la Souris, PHENOMIN-ICS, Illkirch, France
| | - Alain Guimond
- Institut Clinique de la Souris, PHENOMIN-ICS, Illkirch, France
| | - Tania Sorg
- Institut Clinique de la Souris, PHENOMIN-ICS, Illkirch, France
| | - Eric Robinet
- Institute of Image-Guided Surgery, University of Strasbourg, Strasbourg, France
| | - Thomas F Baumert
- Institute of Image-Guided Surgery, University of Strasbourg, Strasbourg, France
| | - Laurent Monassier
- Institut Clinique de la Souris, PHENOMIN-ICS, Illkirch, France.,Laboratoire de Toxicologie et Pharmacologie Neuro Cardiovasculaire, Université de Strasbourg, Strasbourg, France.,Institut National de la Santé et de la Recherche Médicale, Illkirch, France
| | - Yann Herault
- Institut Clinique de la Souris, PHENOMIN-ICS, Illkirch, France.,Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Centre National de la Recherche Scientifique, Illkirch, France
| |
Collapse
|
131
|
El-Kharashi OA, Mohamed DI, Khairy E, Ezzat SF, Zaki WS. Exenatide promotes cardiac lncRNAs HOX transcript antisense RNA (HOTAIR) in Wistar rats with liver cirrhosis; a novel role of GLP-1 receptor agonists in cirrhotic cardiomyopathy. Eur J Pharmacol 2019; 855:294-304. [PMID: 31100415 DOI: 10.1016/j.ejphar.2019.05.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 05/10/2019] [Accepted: 05/13/2019] [Indexed: 11/29/2022]
Abstract
Long acting non-coding RNAs lncRNAs HOX Transcript Antisense RNA (HOTAIR) is cardioprotective and mediates its effect through sirtulin 1 (SIRT1). The decrease in HOTAIR expression predisposes to various types of cardiomyopathy. We aimed to investigate whether decrease HOTAIR expression is involved in cirrhotic cardiomyopathy or not and the role of glucagon like peptide 1 receptor (GLP-1 receptor) in facilitating its effect through studying the effect of a exenatide (EXA), on cardiac function as well as the expression of some relevant bio-molecules. Rats were used and divided into: naïve, EXA, Thioacetamide (TAA) and TAA + EXA groups. ECG, dobutamine stress test (DST) were done. AST, ALT, fasting blood glucose, troponin I were measured. Cardiac HOTAIR & SIRT1, hepatic and cardiac GLP-1 receptor expression levels were investigated in addition to histological studies. Our results showed that EXA administration in control rats produced no significant changes. TAA induced cirrhosis with insulin resistance and significant changes in cardiac functions. GLP-1 receptor, HOTAIR and SIRT1 expression in cardiac tissue were significantly decreased with a significant increase in troponin I. EXA + TAA group showed a restoration of the hepatic architecture and function. EXA treatment produced significant improvement in cardiac parameters and was associated with increasing the expression of cardiac GLP-1 receptor, HOTAIR. The cardiac muscle showed an apparent decrease in collagen fibers. So we can conclude that EXA promotes the protective effect of HOTAIR on cardiac structure and function in rat model of cirrhosis which may introduce a new therapeutic strategy in cirrhotic cardiomyopathy.
Collapse
Affiliation(s)
- Omnyah A El-Kharashi
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| | - Doaa I Mohamed
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| | - Eman Khairy
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| | - Samar F Ezzat
- Department of Histology and Cell Biology, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| | - Walid S Zaki
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
132
|
Ericksen RE, Lim SL, McDonnell E, Shuen WH, Vadiveloo M, White PJ, Ding Z, Kwok R, Lee P, Radda GK, Toh HC, Hirschey MD, Han W. Loss of BCAA Catabolism during Carcinogenesis Enhances mTORC1 Activity and Promotes Tumor Development and Progression. Cell Metab 2019; 29:1151-1165.e6. [PMID: 30661928 PMCID: PMC6506390 DOI: 10.1016/j.cmet.2018.12.020] [Citation(s) in RCA: 181] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 09/13/2018] [Accepted: 12/21/2018] [Indexed: 02/07/2023]
Abstract
Tumors display profound changes in cellular metabolism, yet how these changes aid the development and growth of tumors is not fully understood. Here we use a multi-omic approach to examine liver carcinogenesis and regeneration, and find that progressive loss of branched-chain amino acid (BCAA) catabolism promotes tumor development and growth. In human hepatocellular carcinomas and animal models of liver cancer, suppression of BCAA catabolic enzyme expression led to BCAA accumulation in tumors, though this was not observed in regenerating liver tissues. The degree of enzyme suppression strongly correlated with tumor aggressiveness, and was an independent predictor of clinical outcome. Moreover, modulating BCAA accumulation regulated cancer cell proliferation in vitro, and tumor burden and overall survival in vivo. Dietary BCAA intake in humans also correlated with cancer mortality risk. In summary, loss of BCAA catabolism in tumors confers functional advantages, which could be exploited by therapeutic interventions in certain cancers.
Collapse
Affiliation(s)
- Russell E Ericksen
- Singapore Bioimaging Consortium, Agency for Science, Technology, and Research, 11 Biopolis Way, 138667 Singapore, Singapore
| | - Siew Lan Lim
- Singapore Bioimaging Consortium, Agency for Science, Technology, and Research, 11 Biopolis Way, 138667 Singapore, Singapore
| | - Eoin McDonnell
- Duke Molecular Physiology Institute, 300 North Duke Street, Durham, NC 27701, USA
| | - Wai Ho Shuen
- Division of Medical Oncology, National Cancer Center Singapore, 11 Hospital Drive, 169610 Singapore, Singapore
| | - Maya Vadiveloo
- Department of Nutrition and Food Sciences, University of Rhode Island, 41 Lower College Road, Kingston, RI 02881, USA
| | - Phillip J White
- Duke Molecular Physiology Institute, 300 North Duke Street, Durham, NC 27701, USA
| | - Zhaobing Ding
- Singapore Bioimaging Consortium, Agency for Science, Technology, and Research, 11 Biopolis Way, 138667 Singapore, Singapore
| | - Royston Kwok
- Singapore Bioimaging Consortium, Agency for Science, Technology, and Research, 11 Biopolis Way, 138667 Singapore, Singapore
| | - Philip Lee
- Singapore Bioimaging Consortium, Agency for Science, Technology, and Research, 11 Biopolis Way, 138667 Singapore, Singapore
| | - George K Radda
- Singapore Bioimaging Consortium, Agency for Science, Technology, and Research, 11 Biopolis Way, 138667 Singapore, Singapore
| | - Han Chong Toh
- Division of Medical Oncology, National Cancer Center Singapore, 11 Hospital Drive, 169610 Singapore, Singapore
| | - Matthew D Hirschey
- Duke Molecular Physiology Institute, 300 North Duke Street, Durham, NC 27701, USA
| | - Weiping Han
- Singapore Bioimaging Consortium, Agency for Science, Technology, and Research, 11 Biopolis Way, 138667 Singapore, Singapore.
| |
Collapse
|
133
|
Motawi TMK, Sadik NAH, Sabry D, Shahin NN, Fahim SA. rs2267531, a promoter SNP within glypican-3 gene in the X chromosome, is associated with hepatocellular carcinoma in Egyptians. Sci Rep 2019; 9:6868. [PMID: 31053802 PMCID: PMC6499880 DOI: 10.1038/s41598-019-43376-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 04/23/2019] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a major health concern in Egypt owing to the high prevalence of hepatitis C virus (HCV) infection. HCC incidence is characterized by obvious male predominance, yet the molecular mechanisms behind this gender bias are still unidentified. Functional variations in X-linked genes have more impact on males than females. Glypican-3 (GPC3) gene, located in the Xq26 region, has lately emerged as being potentially implicated in hepatocellular carcinogenesis. The current study was designed to examine the association of -784 G/C single nucleotide polymorphism (SNP) in GPC3 promoter region (rs2267531) with HCC susceptibility in male and female Egyptian HCV patients. Our results revealed a significant association between GPC3 and HCC risk in both males and females, evidenced by higher C allele and CC/C genotype frequencies in HCC patients when compared to controls. However, no such association was found when comparing HCV patients to controls. Moreover, GPC3 gene and protein expression levels were significantly higher in CC/C than in GG/G genotype carriers in males and females. The CC/C genotype exhibited a significant shorter overall survival than GG/G genotype in HCC patients. In conclusion, GPC3 rs2267531 on the X chromosome is significantly associated with HCC, but not with HCV infection, in the Egyptian population.
Collapse
Affiliation(s)
| | | | - Dina Sabry
- Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Nancy Nabil Shahin
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Sally Atef Fahim
- Biochemistry Department, Faculty of Pharmacy, Ahram Canadian University, Cairo, Egypt.
| |
Collapse
|
134
|
Zhang HE, Henderson JM, Gorrell MD. Animal models for hepatocellular carcinoma. Biochim Biophys Acta Mol Basis Dis 2019; 1865:993-1002. [PMID: 31007176 DOI: 10.1016/j.bbadis.2018.08.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 07/31/2018] [Accepted: 08/02/2018] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC) represents ~90% of all cases of primary liver cancer and occurs predominantly in patients with underlying chronic liver disease and cirrhosis. Establishing appropriate animal models for HCC is required for basic and translational studies, especially the models that can recapitulate one of the human disease settings. Current animal models can be categorized as chemically-induced, genetically-engineered, xenograft, or a combination of these with each other or with a metabolic insult. A single approach to resemble human HCC in animals is not sufficient. Combining pathogenic insults in animal models may more realistically recapitulate the multiple etiologic agents occurring in humans. Combining chemical injury with metabolic disorder or alcohol consumption in mice reduces the time taken to hepatocarcinogenesis. Genetically-engineering weak activation of HCC-promoting pathways combined with disease-specific injury models will possibly mimic the pathophysiology of human HCC in distinct clinical settings.
Collapse
Affiliation(s)
- Hui Emma Zhang
- Centenary Institute, The University of Sydney, Newtown, New South Wales, 2042, Australia; The University of Sydney Faculty of Medicine and Health, New South Wales, 2006, Australia
| | - James M Henderson
- Centenary Institute, The University of Sydney, Newtown, New South Wales, 2042, Australia; The University of Sydney Faculty of Medicine and Health, New South Wales, 2006, Australia
| | - Mark D Gorrell
- Centenary Institute, The University of Sydney, Newtown, New South Wales, 2042, Australia; The University of Sydney Faculty of Medicine and Health, New South Wales, 2006, Australia.
| |
Collapse
|
135
|
Loeuillard E, Fischbach SR, Gores GJ, Ilyas SI. Animal models of cholangiocarcinoma. Biochim Biophys Acta Mol Basis Dis 2019; 1865:982-992. [PMID: 29627364 PMCID: PMC6177316 DOI: 10.1016/j.bbadis.2018.03.026] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 03/23/2018] [Accepted: 03/29/2018] [Indexed: 12/18/2022]
Abstract
Cholangiocarcinoma (CCA) is an aggressive biliary tract malignancy with a poor overall prognosis. There is a critical need to develop effective targeted therapies for the treatment of this lethal disease. In an effort to address this challenge, preclinical in vivo studies have become paramount in understanding CCA carcinogenesis, progression, and therapy. Various CCA animal models exist including carcinogen-based models in which animals develop CCA after exposure to a carcinogen, genetically engineered mouse models in which genetic changes are induced in mice leading to CCA, murine syngeneic orthotopic models, as well as xenograft tumors derived from xenotransplantation of CCA cells, organoids, and patient-derived tissue. Each type has distinct advantages as well as shortcomings. In the ideal animal model of CCA, the tumor arises from the biliary tract in an immunocompetent host with a species-matched tumor microenvironment. Such a model would also be time-efficient, recapitulate the genetic and histopathological features of human CCA, and predict therapeutic response in humans. Recently developed biliary tract transduction and orthotopic syngeneic transplant mouse models encompass several of these elements. Herein, we review the different animal models of CCA, their advantages and deficiencies, as well as features which mimic human CCA.
Collapse
Affiliation(s)
- Emilien Loeuillard
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, United States
| | - Samantha R Fischbach
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, United States
| | - Gregory J Gores
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, United States
| | - Sumera I Ilyas
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, United States.
| |
Collapse
|
136
|
Manieri E, Herrera-Melle L, Mora A, Tomás-Loba A, Leiva-Vega L, Fernández DI, Rodríguez E, Morán L, Hernández-Cosido L, Torres JL, Seoane LM, Cubero FJ, Marcos M, Sabio G. Adiponectin accounts for gender differences in hepatocellular carcinoma incidence. J Exp Med 2019; 216:1108-1119. [PMID: 30944152 PMCID: PMC6504215 DOI: 10.1084/jem.20181288] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 01/11/2019] [Accepted: 02/08/2019] [Indexed: 12/24/2022] Open
Abstract
Gender disparity in liver cancer incidence is a relevant feature of this malignancy. Manieri et al. show that testosterone-induced JNK1 activation in adipose tissue results in decreased levels of circulating adiponectin, which is responsible for higher incidence of hepatocellular carcinoma in males. Hepatocellular carcinoma (HCC) is the sixth most common cancer type and the fourth leading cause of cancer-related death. This cancer appears with higher incidence in men and during obesity; however, the specific mechanisms underlying this correlation are unknown. Adipose tissue, a key organ in metabolic syndrome, shows evident gender disparities in the production of adipokines. Levels of the important adipokine adiponectin decrease in men during puberty, as well as in the obese state. Here, we show that this decrease in adiponectin levels is responsible for the increased liver cancer risk in males. We found that testosterone activates the protein JNK in mouse and human adipocytes. JNK-mediated inhibition of adiponectin secretion increases liver cancer cell proliferation, since adiponectin protects against liver cancer development through the activation of AMP-activated protein kinase (AMPK) and p38α. This study provides insight into adipose tissue to liver crosstalk and its gender relation during cancer development, having the potential to guide strategies for new cancer therapeutics.
Collapse
Affiliation(s)
- Elisa Manieri
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Centro Nacional de Biotecnología, Madrid, Spain
| | | | - Alfonso Mora
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Antonia Tomás-Loba
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Luis Leiva-Vega
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Delia I Fernández
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Elena Rodríguez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Laura Morán
- Department of Immunology, Ophthalmology and Otorhinolaryngology, Complutense University School of Medicine, Madrid, Spain.,12 de Octubre Health Research Institute, Madrid, Spain
| | - Lourdes Hernández-Cosido
- University of Salamanca, University Hospital of Salamanca-Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain
| | - Jorge L Torres
- University of Salamanca, University Hospital of Salamanca-Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain
| | - Luisa M Seoane
- Fisiopatología Endocrina, Instituto de Investigación Sanitaria de Santiago, Hospital Clínico Universitario de Santiago de Compostela Servicio Gallego de Salud, Santiago de Compostela, Spain.,Centro de Investigación Biomédica en Red (CIBER), Fisiopatología Obesidad y Nutrición, Instituto Salud Carlos III, Spain
| | - Francisco Javier Cubero
- Department of Immunology, Ophthalmology and Otorhinolaryngology, Complutense University School of Medicine, Madrid, Spain.,12 de Octubre Health Research Institute, Madrid, Spain
| | - Miguel Marcos
- University of Salamanca, University Hospital of Salamanca-Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain
| | - Guadalupe Sabio
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| |
Collapse
|
137
|
El-Kharrag R, Owen R, Boison D. Adenosine Kinase Deficiency Increases Susceptibility to a Carcinogen. J Caffeine Adenosine Res 2019; 9:4-11. [PMID: 30944910 DOI: 10.1089/caff.2018.0019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Background: Adenosine kinase (ADK) is a key regulator of hepatic metabolism. Its deficiency in the liver causes hepatic steatosis and methylation defects. In this study, we investigated whether reduced ADK expression affects the susceptibility of the liver to a carcinogen. Methods: We investigated ADK expression in samples from 11 liver cancer patients. We used transgenic Adk-tg mice with reduced hepatic ADK to study their susceptibility to a carcinogen. We exposed 45 Adk-tg and 21 wild-type (WT) mice to the carcinogen diethylnitrosamine (DEN) and the tumor promoter phenobarbital (PB) and examined the survival and body weight. Results: Seven of 11 patients with liver cancer had reduced ADK expression. A Kaplan-Meier survival curve showed a significantly increased mortality rate of DEN/PB-exposed Adk-tg mice compared with WT mice. Conclusions: Reduced hepatic ADK increases the susceptibility to the acute toxic effects of a carcinogen. Low hepatic ADK might be a risk factor and biomarker for cancer development.
Collapse
Affiliation(s)
- Rkia El-Kharrag
- Department of Neurobiology, Legacy Research Institute, Portland, Oregon
| | - Randy Owen
- Department of Neurobiology, Legacy Research Institute, Portland, Oregon
| | - Detlev Boison
- Department of Neurobiology, Legacy Research Institute, Portland, Oregon
| |
Collapse
|
138
|
Jiang J, Chen Y, Dong T, Yue M, Zhang Y, An T, Zhang J, Liu P, Yang X. Polydatin inhibits hepatocellular carcinoma via the AKT/STAT3-FOXO1 signaling pathway. Oncol Lett 2019; 17:4505-4513. [PMID: 30944640 PMCID: PMC6444395 DOI: 10.3892/ol.2019.10123] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 01/15/2019] [Indexed: 12/13/2022] Open
Abstract
Polydatin, extracted from Polygonum cuspidatum, is known for its anti-platelet aggregation and anti-inflammatory effects. However, studies on the association of polydatin with cancer are limited, particularly with regards to epithelial-mesenchymal transition (EMT)-associated migration and invasion of cancer cells. The purpose of the present study was to reveal the potential anticancer effects of polydatin on hepatocellular carcinoma (HCC) cells, particularly its effects on EMT. MTT assay was used to determine cell viability. Migration and invasion were evaluated through wound healing and transwell assays. Colony formation efficiency assay was conducted to detect proliferation. Flow cytometric analyses of apoptosis and cell cycle progression were performed following cells staining with Annexin V-fluorescein isothiocyanate (FITC)/propidium iodide (PI) and PI alone, respectively. Western blotting was used to investigate relevant molecular mechanisms. The results indicated that polydatin inhibited proliferation via G2/M arrest, suppressed migration and invasion of HCC cells, and promoted their apoptosis. In addition, phosphorylated (p)-protein kinase B (AKT), p-Janus kinase 1 and p-signal transducer and activator of transcription 3 (STAT3) levels were decreased as polydatin concentrations increased, and forkhead box protein O1 (FOXO1) expression was upregulated. Furthermore, the expression levels of various markers of EMT were reversed following treatment with polydatin. In conclusion, the present study validated that polydatin may inhibit proliferation via G2/M arrest, and suppressed EMT-associated migration and invasion of HCC cells. The results also suggested that polydatin may promote HCC cell apoptosis by blocking the AKT/STAT3-FOXO1 signaling pathway.
Collapse
Affiliation(s)
- Jian Jiang
- Department of Abdominal Ultrasonography, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Yaodong Chen
- Department of Abdominal Ultrasonography, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Tianxiu Dong
- Department of Abdominal Ultrasonography, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Minlu Yue
- Department of Abdominal Ultrasonography, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Yu Zhang
- Department of Abdominal Ultrasonography, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Tingting An
- Department of Abdominal Ultrasonography, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Jiuwei Zhang
- Department of Abdominal Ultrasonography, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Pengfei Liu
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Xiuhua Yang
- Department of Abdominal Ultrasonography, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
139
|
Callegari E, Domenicali M, Shankaraiah RC, D'Abundo L, Guerriero P, Giannone F, Baldassarre M, Bassi C, Elamin BK, Zagatti B, Ferracin M, Fornari F, Altavilla G, Blandamura S, Silini EM, Gramantieri L, Sabbioni S, Negrini M. MicroRNA-Based Prophylaxis in a Mouse Model of Cirrhosis and Liver Cancer. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 14:239-250. [PMID: 30641476 PMCID: PMC6330511 DOI: 10.1016/j.omtn.2018.11.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 11/26/2018] [Accepted: 11/26/2018] [Indexed: 02/07/2023]
Abstract
Most hepatocellular carcinomas (HCCs) arise in the context of chronic liver disease and/or cirrhosis. Thus, chemoprevention in individuals at risk represents an important but yet unproven approach. In this study, we investigated the ability of microRNA (miRNA)-based molecules to prevent liver cancer development in a cirrhotic model. To this end, we developed a mouse model able to recapitulate the natural progression from fibrosis to HCC, and then we tested the prophylactic activity of an miRNA-based approach in the model. The experiments were carried out in the TG221 transgenic mouse, characterized by the overexpression of miR-221 in the liver and predisposed to the development of liver tumors. TG221 as well as wild-type mice were exposed to the hepatotoxin carbon tetrachloride (CCl4) to induce chronic liver damage. All mice developed liver cirrhosis, but only TG221 mice developed nodular lesions in 100% of cases within 6 months of age. The spectrum of lesions ranged from dysplastic foci to carcinomas. To investigate miRNA-based prophylactic approaches, anti-miR-221 oligonucleotides or miR-199a-3p mimics were administered to TG221 CCl4-treated mice. Compared to control animals, a significant reduction in number, size, and, most significantly, malignant phenotype of liver nodules was observed, thus demonstrating an important prophylactic action of miRNA-based molecules. In summary, in this article, we not only report a simple model of liver cancer in a cirrhotic background but also provide evidence for a potential miRNA-based approach to reduce the risk of HCC development.
Collapse
Affiliation(s)
- Elisa Callegari
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | - Marco Domenicali
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy; Center for Applied Biomedical Research, St. Orsola-Malpighi University Hospital, 40138 Bologna, Italy
| | - Ram Charan Shankaraiah
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Lucilla D'Abundo
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Paola Guerriero
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Ferdinando Giannone
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy; Center for Applied Biomedical Research, St. Orsola-Malpighi University Hospital, 40138 Bologna, Italy
| | - Maurizio Baldassarre
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy; Center for Applied Biomedical Research, St. Orsola-Malpighi University Hospital, 40138 Bologna, Italy
| | - Cristian Bassi
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Bahaeldin K Elamin
- Department of Basic Sciences, College of Medicine, University of Bisha, 61922 Bisha, Saudi Arabia; Department of Medical Microbiology, Faculty of Medical Laboratory Sciences, University of Khartoum, 11115 Khartoum, Sudan
| | - Barbara Zagatti
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Manuela Ferracin
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy
| | - Francesca Fornari
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy; Center for Applied Biomedical Research, St. Orsola-Malpighi University Hospital, 40138 Bologna, Italy
| | | | - Stella Blandamura
- Department of Medicine DIMED, University of Padova, 35121 Padova, Italy
| | - Enrico Maria Silini
- Section of Anatomy and Pathology, University Hospital of Parma, 43121 Parma, Italy
| | - Laura Gramantieri
- Center for Applied Biomedical Research, St. Orsola-Malpighi University Hospital, 40138 Bologna, Italy
| | - Silvia Sabbioni
- Department of Life Sciences and Biotechnologies, University of Ferrara, 44121 Ferrara, Italy
| | - Massimo Negrini
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| |
Collapse
|
140
|
Arboatti AS, Lambertucci F, Sedlmeier MG, Pisani G, Monti J, Álvarez MDL, Francés DEA, Ronco MT, Carnovale CE. Diethylnitrosamine enhances hepatic tumorigenic pathways in mice fed with high fat diet (Hfd). Chem Biol Interact 2019; 303:70-78. [PMID: 30826251 DOI: 10.1016/j.cbi.2019.02.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 01/28/2019] [Accepted: 02/24/2019] [Indexed: 12/21/2022]
Abstract
Obesity has been implicated in the genesis of metabolic syndromes including insulin resistance and Type 2 Diabetes Mellitus (T2DM). Given the association between T2DM and the risk of hepatocellular carcinoma (HCC), our specific goal was to determine whether the liver of HFD-induced T2DM mice is more sensitive to the carcinogen diethylnitrosamine (DEN), due to a modification of the molecular pathways implicated in the early stages of HCC pathogenesis. C57BL/6 male mice (five-week-old) were divided into 4 groups: C, C + DEN, HFD and HFD + DEN. Mice were euthanized twenty-five weeks after DEN-injection. Livers of HDF-fed mice showed a higher proliferative index than Control groups. In line with this, HFD groups showed an increase of nuclear β-catenin, and interestingly, DEN treatment led to a slight increase in the expression of this protein in HFD group. Based on these results, and to confirm this effect, we analyzed β-catenin target genes, finding that DEN treatment in HFD group led to a significant increase of Vegf, c-myc, c-jun and cyclin D1 expression levels. According to our results, the expression of TCF4 showed to be significantly increased in HFD + DEN vs. HFD. In this regard, the β-catenin/TCF4 complex enhanced its association with pSmads 2/3, as we observed an increase of nuclear Smads expression in HFD + DEN, suggesting a possible role of TGF-β1/Smads signaling pathway in this phenomenon. Our results show that the liver of HFD fed model that resembles early T2DM pathology in mice, is more sensitive to DEN, by inducing both Wnt/β-catenin and TGF β1/Smads tumorigenic pathways.
Collapse
Affiliation(s)
- A S Arboatti
- Instituto de Fisiología Experimental (IFISE-CONICET), Cátedra de Fisiología, Facultad de Ciencias Bioquímicas y Farmacéuticas- UNR, Suipacha 570, 2000, Rosario, Argentina
| | - F Lambertucci
- Instituto de Fisiología Experimental (IFISE-CONICET), Cátedra de Fisiología, Facultad de Ciencias Bioquímicas y Farmacéuticas- UNR, Suipacha 570, 2000, Rosario, Argentina
| | - M G Sedlmeier
- Instituto de Fisiología Experimental (IFISE-CONICET), Cátedra de Fisiología, Facultad de Ciencias Bioquímicas y Farmacéuticas- UNR, Suipacha 570, 2000, Rosario, Argentina
| | - G Pisani
- Cátedra de Morfología, Facultad de Ciencias Bioquímicas y Farmacéuticas, UNR, Suipacha 570, 2000, Rosario, Argentina
| | - J Monti
- Instituto de Fisiología Experimental (IFISE-CONICET), Cátedra de Fisiología, Facultad de Ciencias Bioquímicas y Farmacéuticas- UNR, Suipacha 570, 2000, Rosario, Argentina
| | - M de L Álvarez
- Instituto de Fisiología Experimental (IFISE-CONICET), Cátedra de Fisiología, Facultad de Ciencias Bioquímicas y Farmacéuticas- UNR, Suipacha 570, 2000, Rosario, Argentina; Cátedra de Morfología, Facultad de Ciencias Bioquímicas y Farmacéuticas, UNR, Suipacha 570, 2000, Rosario, Argentina
| | - D E A Francés
- Instituto de Fisiología Experimental (IFISE-CONICET), Cátedra de Fisiología, Facultad de Ciencias Bioquímicas y Farmacéuticas- UNR, Suipacha 570, 2000, Rosario, Argentina
| | - M T Ronco
- Instituto de Fisiología Experimental (IFISE-CONICET), Cátedra de Fisiología, Facultad de Ciencias Bioquímicas y Farmacéuticas- UNR, Suipacha 570, 2000, Rosario, Argentina
| | - C E Carnovale
- Instituto de Fisiología Experimental (IFISE-CONICET), Cátedra de Fisiología, Facultad de Ciencias Bioquímicas y Farmacéuticas- UNR, Suipacha 570, 2000, Rosario, Argentina.
| |
Collapse
|
141
|
Wang Q, Zhang P, Li Z, Feng X, Lv C, Zhang H, Xiao H, Ding J, Chen X. Evaluation of Polymer Nanoformulations in Hepatoma Therapy by Established Rodent Models. Theranostics 2019; 9:1426-1452. [PMID: 30867842 PMCID: PMC6401493 DOI: 10.7150/thno.31683] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 01/08/2019] [Indexed: 01/10/2023] Open
Abstract
Hepatoma is one of the most severe malignancies usually with poor prognosis, and many patients are insensitive to the existing therapeutic agents, including the drugs for chemotherapy and molecular targeted therapy. Currently, researchers are committed to developing the advanced formulations with controlled drug delivery to improve the efficacy of hepatoma therapy. Numerous inoculated, induced, and genetically engineered hepatoma rodent models are now available for formulation screening. However, animal models of hepatoma cannot accurately represent human hepatoma in terms of histological characteristics, metastatic pathways, and post-treatment responses. Therefore, advanced animal hepatoma models with comparable pathogenesis and pathological features are in urgent need in the further studies. Moreover, the development of nanomedicines has renewed hope for chemotherapy and molecular targeted therapy of advanced hepatoma. As one kind of advanced formulations, the polymer-based nanoformulated drugs have many advantages over the traditional ones, such as improved tumor selectivity and treatment efficacy, and reduced systemic side effects. In this article, the construction of rodent hepatoma model and much information about the current development of polymer nanomedicines were reviewed in order to provide a basis for the development of advanced formulations with clinical therapeutic potential for hepatoma.
Collapse
Affiliation(s)
- Qilong Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun 130021, P. R. China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
| | - Ping Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun 130021, P. R. China
| | - Zhongmin Li
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, P. R. China
| | - Xiangru Feng
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, P. R. China
| | - Chengyue Lv
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, P. R. China
| | - Huaiyu Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, P. R. China
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, P. R. China
| |
Collapse
|
142
|
Mo'men YS, Hussein RM, Kandeil MA. Involvement of PI3K/Akt pathway in the protective effect of hesperidin against a chemically induced liver cancer in rats. J Biochem Mol Toxicol 2019; 33:e22305. [PMID: 30779474 DOI: 10.1002/jbt.22305] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 11/14/2018] [Accepted: 01/09/2019] [Indexed: 12/14/2022]
Abstract
Hesperidin is a flavanone glycoside that is found in the Citrus species and showed antioxidant, hepatoprotective as well as anticancer activity. This study investigated the effect of hesperidin on the PI3K/Akt pathway as a possible mechanism for its protective effect against diethylnitrosamine (DEN)-induced hepatocellular carcinoma (HCC). Adult Wistar rats were divided into Control group (received drug vehicle); DEN group (received 100 mg/L of DEN solution for 8 weeks), and hesperidin + DEN group (received 200 mg/kg body weight of hesperidin/day orally for 16 weeks + DEN solution as DEN group). Our findings showed that the administration of hesperidin significantly decreased the elevation in liver function enzymes, serum AFP level, and oxidative stress markers. Moreover, hesperidin administration suppressed DEN-induced upregulation of PI3K, Akt, CDK-2 protein expression, and preserved the integrity of the liver tissues from HCC formation. In conclusion, the hepatoprotective activity of hesperidin is mediated via its antioxidation and downregulation of the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Yomna S Mo'men
- Department of Biochemistry, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Rasha M Hussein
- Department of Biochemistry, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Mohamed A Kandeil
- Department of Biochemistry, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, Egypt
| |
Collapse
|
143
|
WDR76 is a RAS binding protein that functions as a tumor suppressor via RAS degradation. Nat Commun 2019; 10:295. [PMID: 30655611 PMCID: PMC6336889 DOI: 10.1038/s41467-018-08230-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 12/19/2018] [Indexed: 12/15/2022] Open
Abstract
Stability regulation of RAS that can affect its activity, in addition to the oncogenic mutations, occurs in human cancer. However, the mechanisms for stability regulation of RAS involved in their activity and its roles in tumorigenesis are poorly explored. Here, we identify WD40-repeat protein 76 (WDR76) as one of the HRAS binding proteins using proteomic analyses of hepatocellular carcinomas (HCC) tissue. WDR76 plays a role as an E3 linker protein and mediates the polyubiquitination-dependent degradation of RAS. WDR76-mediated RAS destabilization results in the inhibition of proliferation, transformation, and invasion of liver cancer cells. WDR76-/- mice are more susceptible to diethylnitrosamine-induced liver carcinogenesis. Liver-specific WDR76 induction destabilizes Ras and markedly reduces tumorigenesis in HRasG12V mouse livers. The clinical relevance of RAS regulation by WDR76 is indicated by the inverse correlation of their expressions in HCC tissues. Our study demonstrates that WDR76 functions as a tumor suppressor via RAS degradation.
Collapse
|
144
|
Pioglitazone Reduces Hepatocellular Carcinoma Development in Two Rodent Models of Cirrhosis. J Gastrointest Surg 2019; 23:101-111. [PMID: 30367397 PMCID: PMC6328630 DOI: 10.1007/s11605-018-4004-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 10/05/2018] [Indexed: 01/31/2023]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is one of the deadliest malignancies worldwide due to the lack of effective treatments. Chemoprevention in high-risk patients is a promising, alternative strategy. In this study, pioglitazone was investigated for its ability to prevent hepatocarcinogenesis in two rodent models of cirrhosis. METHODS In the first model, male Wistar rats were given repeated, low-dose injections of diethylnitrosamine (DEN) to accurately recapitulate the progression of fibrosis to cirrhosis and HCC. In the second model, a single dose of DEN was administered to male C57Bl/6 pups at day fifteen followed by administration of a choline-deficient, L-amino acid defined, high-fat diet (CDAHFD) at week six for 24 weeks. Pioglitazone treatment started at the first signs of fibrosis in both models. RESULTS Pioglitazone effectively reduced fibrosis progression and HCC development in both models. Gross tumor nodules were significantly reduced after pioglitazone treatment (7.4 ± 1.6 vs. 16.6 ± 2.6 in the rat DEN model and 5.86 ± 1.82 vs. 13.2 ± 1.25 in the mouse DEN+CDAHFD model). In both models, pioglitazone reduced the activation of mitogen-activated protein kinase (MAPK) and upregulated the hepato-protective AMP-activated protein kinase (AMPK) pathway via increasing circulating adiponectin production. CONCLUSION Pioglitazone is an effective agent for chemoprevention in rodents and could be repurposed as a multi-targeted drug for delaying liver fibrosis and hepatocarcinogenesis.
Collapse
|
145
|
Yamamoto M, Xin B, Nishikawa Y. Mouse Model for Hepatocellular Carcinoma and Cholangiocarcinoma Originated from Mature Hepatocytes. Methods Mol Biol 2019; 1905:221-236. [PMID: 30536104 DOI: 10.1007/978-1-4939-8961-4_20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Liver cancer consists of two main histological subtypes, hepatocellular carcinoma and cholangiocarcinoma, both of which have poor prognosis. Therefore, in searching for new therapeutic targets, adequate mouse models to develop and validate therapeutic strategies are urgently needed. Although there are mouse models of liver cancer, each model has shortcomings. To overcome these shortcomings, a mouse model using a hydrodynamic tail vein injection and the Sleeping Beauty transposon was developed. By inducing stable expression of oncogenes in mouse hepatocytes in vivo, the model can easily induce liver cancer with specific characteristics that depend on the oncogenes used to induce carcinogenesis. Here, we describe the details of the methods to induce hepatocellular carcinoma or cholangiocarcinoma from mouse hepatocytes.
Collapse
Affiliation(s)
- Masahiro Yamamoto
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University, Asahikawa, Hokkaido, Japan.
| | - Bing Xin
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Yuji Nishikawa
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| |
Collapse
|
146
|
Liu YT, Tseng TC, Soong RS, Peng CY, Cheng YH, Huang SF, Chuang TH, Kao JH, Huang LR. A novel spontaneous hepatocellular carcinoma mouse model for studying T-cell exhaustion in the tumor microenvironment. J Immunother Cancer 2018; 6:144. [PMID: 30526672 PMCID: PMC6286542 DOI: 10.1186/s40425-018-0462-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 11/23/2018] [Indexed: 12/15/2022] Open
Abstract
Immunotherapy has ushered in a new era of cancer therapy, and this is also applicable to therapy of hepatocellular carcinoma (HCC). In this context, effective development of therapeutic strategies requires an HCC mouse model with known tumor-associated antigens (TAAs) and an HCC growth reporter. We created such a model using hydrodynamic injection and a transposon system to introduce AKT and NRAS and open reading frames (ORFs) encoding surrogate tumor antigens and luciferase into chromosomes of hepatocytes to induce nodular and diffuse tumors in the liver. TAA-specific CD8+ T cells were detected during HCC progression; however, these showed exhausted-like phenotypes and were unable to control tumor growth. Myeloid-derived suppressor cells (MDSCs) and tumor-associated macrophages (TAM) from the tumor microenvironment were found to contribute to the suppression of the CD8+ T-cell response. The transposon-based Akt/N-Ras-induced HCC mouse model we developed enables researchers to monitor tumor growth non-invasively and to quantify and characterize endogenous or adoptively transferred TAA-specific CD8+ T-cell responses. These features make it a suitable preclinical model for exploration and evaluation of immune checkpoint inhibitors and cell-based immunotherapies for HCC treatment.
Collapse
Affiliation(s)
- Yu-Tzu Liu
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, No.35, Keyan Road, Zhunan Town, Miaoli County, 350, Taiwan
| | - Tai-Chung Tseng
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Hepatitis Research Center, National Taiwan University, National Taiwan University Hospital, Taipei, Taiwan
| | - Ruey-Shyang Soong
- Department of General Surgery, Chang Gung Memorial Hospital, Keelung, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chun-Yi Peng
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, No.35, Keyan Road, Zhunan Town, Miaoli County, 350, Taiwan
| | - Yu-Hsing Cheng
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, No.35, Keyan Road, Zhunan Town, Miaoli County, 350, Taiwan
| | - Shiu-Feng Huang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, No.35, Keyan Road, Zhunan Town, Miaoli County, 350, Taiwan
| | - Tsung-Hsien Chuang
- Immunology Research Center, National Health Research Institutes, Miaoli, Taiwan
| | - Jia-Horng Kao
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Hepatitis Research Center, National Taiwan University, National Taiwan University Hospital, Taipei, Taiwan.,Graduate Institute of Clinical Medicine, National Taiwan University, Taipei, Taiwan.,Department of Medical Research, National Taiwan University, National Taiwan University Hospital, Taipei, Taiwan
| | - Li-Rung Huang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, No.35, Keyan Road, Zhunan Town, Miaoli County, 350, Taiwan. .,Graduate Institute of Clinical Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
147
|
Tripathy A, Thakurela S, Sahu MK, Uthanasingh K, Behera M, Ajay AK, Kumari R. The molecular connection of histopathological heterogeneity in hepatocellular carcinoma: A role of Wnt and Hedgehog signaling pathways. PLoS One 2018; 13:e0208194. [PMID: 30513115 PMCID: PMC6279049 DOI: 10.1371/journal.pone.0208194] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 11/13/2018] [Indexed: 12/21/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is leading cause of cancer-related mortality and is categorized among the most common malignancies around the world. It is a heterogeneous tumor, which shows significant degree of histopathological heterogeneity. Despite the apparent histopathological diversity there has been very little distinct correlation between histopathological features and molecular aberrations particularly when it comes to the expression level of Wnt and Hh pathway molecules. The role of Wnt and Hh pathways in relation to HCC behavior viz. histopathological heterogeneity and aggressiveness is not known. Determining the sequential molecular changes and associated histopathological characteristic during HCC initiation, promotion, and progression would probably lead to a better treatment and prognosis. Methods N-Nitrosodiethylamine (DEN) induced HCC model in male Wistar rats were established to study the expression level of Wnt and Hh pathway molecules during different stages of hepatocarcinogenesis. Their expression levels were checked at mRNA and protein levels at initiation, promotion, and progression stages of HCC. The expression levels of Wnt and Hh pathway molecules were correlated with biospecimens of HCC patients of different stages. Results In the present study we identified the comprehensive change in the expression pattern of Wnt and Hh pathway molecules in DEN induced rodent hepatocarcinogenesis model. Our results demonstrate that β-catenin /CTNNB1 plays important role in tumor initiation and promotion by stimulating tumor cell proliferation. The activated Wnt signaling in early stage of HCC is associated with well-differentiated histological pattern. The Hh activity although activated during the initiation stage but is significantly increased during the early promotion stage of hepatocarcinogenesis. The increased activity of both Wnt & Hh pathways during promotion stage is associated with moderately-differentiated histological pattern and was simultaneously linked with an increased expression of MMP9. Furthermore, our data demonstrated that during the progression stage Wnt pathway is modestly down-regulated but the Hh pathway activity sustained which in turn is associated with aggressive and invasive phenotype and poorly-differentiated histopathology. Conclusion Our data uncovers the grade related expression of Wnt and Hh pathway molecules and the potential utility of these molecular signatures in daily clinical practice is to decide best therapy according to patients characteristic. Additionally, our data offer insight into the interaction between Wnt and Hh pathways which triggers HCC development and progression.
Collapse
Affiliation(s)
- Anindita Tripathy
- Disease Biology Lab, KIIT School of Biotechnology, KIIT University, Bhubaneswar, India
| | - Sudhir Thakurela
- Broad Institute of MIT and Harvard, Cambridge, MA, United States of America
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, United States of America
| | - Manoj Kumar Sahu
- Department of Gastroenterology & Hepatobiliary Sciences, IMS & SUM Hospital, Bhubaneswar, India
| | - Kanishka Uthanasingh
- Department of Gastroenterology & Hepatobiliary Sciences, IMS & SUM Hospital, Bhubaneswar, India
| | - Manas Behera
- Department of Gastroenterology & Hepatobiliary Sciences, IMS & SUM Hospital, Bhubaneswar, India
| | - Amrendra Kumar Ajay
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Ratna Kumari
- Disease Biology Lab, KIIT School of Biotechnology, KIIT University, Bhubaneswar, India
- * E-mail:
| |
Collapse
|
148
|
Lee C, Kim M, Lee JH, Oh J, Shin HH, Lee SM, Scherer PE, Kwon HM, Choi JH, Park J. COL6A3-derived endotrophin links reciprocal interactions among hepatic cells in the pathology of chronic liver disease. J Pathol 2018; 247:99-109. [PMID: 30246318 DOI: 10.1002/path.5172] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 08/23/2018] [Accepted: 09/12/2018] [Indexed: 12/22/2022]
Abstract
Extracellular matrix dysregulation is associated with chronic liver disease. CollagenVI-alpha3 chain (COL6A3) is a biomarker for hepatic fibrosis and poor prognosis of hepatocellular carcinoma (HCC), but its function in liver pathology remains unknown. High levels of COL6A3 and its cleaved product, endotrophin (ETP) in tumor-neighboring regions are strongly associated with poor prognosis in HCC patients. Here, we report that the high levels of ETP in injured hepatocytes induce JNK-dependent hepatocyte apoptosis and activate nonparenchymal cells to lead further activation of hepatic inflammation, fibrosis, and apoptosis. Nevertheless ETP per se showed limited phenotypic changes in normal liver tissues. Furthermore, inhibition of ETP activity by utilizing neutralizing antibodies efficiently suppressed the pathological consequences in chronic liver diseases. Our results implicate ETP mechanistically as a crucial mediator in reciprocal interactions among various hepatic cell populations in the pathogenesis of chronic liver disease, and it could be a promising therapeutic target particularly in individuals with high local levels of COL6A3. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Changhu Lee
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, South Korea
| | - Min Kim
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, South Korea.,National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, South Korea
| | - Jun Ho Lee
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, South Korea
| | - Jiyoung Oh
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, South Korea
| | - Hyun-Hee Shin
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, South Korea
| | - Sang Min Lee
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, South Korea
| | - Philipp E Scherer
- Touchstone Diabetes Center, Departments of Internal Medicine and Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Hyug Moo Kwon
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, South Korea
| | - Jang Hyun Choi
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, South Korea
| | - Jiyoung Park
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, South Korea
| |
Collapse
|
149
|
Dow M, Pyke RM, Tsui BY, Alexandrov LB, Nakagawa H, Taniguchi K, Seki E, Harismendy O, Shalapour S, Karin M, Carter H, Font-Burgada J. Integrative genomic analysis of mouse and human hepatocellular carcinoma. Proc Natl Acad Sci U S A 2018; 115:E9879-E9888. [PMID: 30287485 PMCID: PMC6196518 DOI: 10.1073/pnas.1811029115] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cancer genomics has enabled the exhaustive molecular characterization of tumors and exposed hepatocellular carcinoma (HCC) as among the most complex cancers. This complexity is paralleled by dozens of mouse models that generate histologically similar tumors but have not been systematically validated at the molecular level. Accurate models of the molecular pathogenesis of HCC are essential for biomedical progress; therefore we compared genomic and transcriptomic profiles of four separate mouse models [MUP transgenic, TAK1-knockout, carcinogen-driven diethylnitrosamine (DEN), and Stelic Animal Model (STAM)] with those of 987 HCC patients with distinct etiologies. These four models differed substantially in their mutational load, mutational signatures, affected genes and pathways, and transcriptomes. STAM tumors were most molecularly similar to human HCC, with frequent mutations in Ctnnb1, similar pathway alterations, and high transcriptomic similarity to high-grade, proliferative human tumors with poor prognosis. In contrast, TAK1 tumors better reflected the mutational signature of human HCC and were transcriptionally similar to low-grade human tumors. DEN tumors were least similar to human disease and almost universally carried the Braf V637E mutation, which is rarely found in human HCC. Immune analysis revealed that strain-specific MHC-I genotype can influence the molecular makeup of murine tumors. Thus, different mouse models of HCC recapitulate distinct aspects of HCC biology, and their use should be adapted to specific questions based on the molecular features provided here.
Collapse
Affiliation(s)
- Michelle Dow
- Division of Medical Genetics, Department of Medicine, University of California, San Diego, La Jolla, CA 92093
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, La Jolla, CA 92093
- Health Science, Department of Biomedical Informatics, School of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Rachel M Pyke
- Division of Medical Genetics, Department of Medicine, University of California, San Diego, La Jolla, CA 92093
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, La Jolla, CA 92093
| | - Brian Y Tsui
- Division of Medical Genetics, Department of Medicine, University of California, San Diego, La Jolla, CA 92093
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, La Jolla, CA 92093
| | - Ludmil B Alexandrov
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093
| | - Hayato Nakagawa
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 113-8655 Tokyo, Japan
| | - Koji Taniguchi
- Laboratory of Gene Regulation and Signal Transduction, School of Medicine, University of California, San Diego, La Jolla, CA 92093
- Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA 92093
- Department of Pathology, School of Medicine, University of California, San Diego, La Jolla, CA 92093
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Ekihiro Seki
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Olivier Harismendy
- Health Science, Department of Biomedical Informatics, School of Medicine, University of California, San Diego, La Jolla, CA 92093
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Shabnam Shalapour
- Laboratory of Gene Regulation and Signal Transduction, School of Medicine, University of California, San Diego, La Jolla, CA 92093
- Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA 92093
- Department of Pathology, School of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, School of Medicine, University of California, San Diego, La Jolla, CA 92093;
- Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA 92093
- Department of Pathology, School of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Hannah Carter
- Division of Medical Genetics, Department of Medicine, University of California, San Diego, La Jolla, CA 92093;
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093
- Cancer Cell Map Initiative, University of California, San Diego, La Jolla, CA 92093
| | - Joan Font-Burgada
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA 19111
| |
Collapse
|
150
|
Martínez-Cardona C, Lozano-Ruiz B, Bachiller V, Peiró G, Algaba-Chueca F, Gómez-Hurtado I, Such J, Zapater P, Francés R, González-Navajas JM. AIM2 deficiency reduces the development of hepatocellular carcinoma in mice. Int J Cancer 2018; 143:2997-3007. [PMID: 30133699 DOI: 10.1002/ijc.31827] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 07/10/2018] [Accepted: 08/13/2018] [Indexed: 12/26/2022]
Abstract
Chronic liver inflammation is crucial in the pathogenesis of hepatocellular carcinoma (HCC). Activation of the inflammasome complex is a key inflammatory process that has been associated with different liver diseases, but its role in HCC development remains largely unexplored. Here we analyzed the impact of different inflammasome components, including absent in melanoma 2 (AIM2) and NOD-like receptor family pyrin domain containing 3 (NLRP3), in the development of diethylnitrosamine (DEN)-induced HCC in mice. Genetic inactivation of AIM2, but not NLRP3, reduces liver damage and HCC development in this model. AIM2 deficiency ameliorates inflammasome activation, liver inflammation and proliferative responses during HCC initiation. We also identified that AIM2 is highly expressed in Kupffer cells, and that AIM2-mediated production of IL-1β by these cells is enhanced after DEN-induced liver damage. Our data indicate that AIM2 promotes inflammation during carcinogenic liver injury and that it contributes to genotoxic HCC development in mice, thereby recognizing AIM2 as a potential therapeutic target in this disease.
Collapse
Affiliation(s)
- Claudia Martínez-Cardona
- Alicante Institute for Health and Biomedical Research (ISABIAL-FISABIO), Hospital General Universitario de Alicante, Alicante, Spain.,Biomedical Research Network for Hepatic and Digestive Diseases (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain.,Department of Pharmacology, University Miguel Hernández, Elche, Spain
| | - Beatriz Lozano-Ruiz
- Alicante Institute for Health and Biomedical Research (ISABIAL-FISABIO), Hospital General Universitario de Alicante, Alicante, Spain.,Biomedical Research Network for Hepatic and Digestive Diseases (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Victoria Bachiller
- Biomedical Research Network for Hepatic and Digestive Diseases (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Gloria Peiró
- Alicante Institute for Health and Biomedical Research (ISABIAL-FISABIO), Hospital General Universitario de Alicante, Alicante, Spain.,Pathology Department, Hospital General Universitario de Alicante, Alicante, Spain
| | - Francisco Algaba-Chueca
- Biomedical Research Network for Hepatic and Digestive Diseases (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Isabel Gómez-Hurtado
- Alicante Institute for Health and Biomedical Research (ISABIAL-FISABIO), Hospital General Universitario de Alicante, Alicante, Spain.,Biomedical Research Network for Hepatic and Digestive Diseases (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - José Such
- Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Pedro Zapater
- Alicante Institute for Health and Biomedical Research (ISABIAL-FISABIO), Hospital General Universitario de Alicante, Alicante, Spain.,Biomedical Research Network for Hepatic and Digestive Diseases (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain.,Department of Pharmacology, University Miguel Hernández, Elche, Spain
| | - Rubén Francés
- Alicante Institute for Health and Biomedical Research (ISABIAL-FISABIO), Hospital General Universitario de Alicante, Alicante, Spain.,Biomedical Research Network for Hepatic and Digestive Diseases (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain.,Department of Clinical Medicine, University Miguel Hernández, Elche, Spain
| | - José Manuel González-Navajas
- Alicante Institute for Health and Biomedical Research (ISABIAL-FISABIO), Hospital General Universitario de Alicante, Alicante, Spain.,Biomedical Research Network for Hepatic and Digestive Diseases (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain.,The Second Affiliated Hospital, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffman Institute, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|