1
|
Alzahrani AK, Khan A, Singla N, Hai A, Alzahrani AR, Kamal M, Asdaq SMB, Alsalman AJ, Hawaj MAA, Al Odaini LH, Dzinamarira T, Imran M. From diagnosis to therapy: The critical role of lncRNAs in hepatoblastoma. Pathol Res Pract 2024; 260:155412. [PMID: 38889493 DOI: 10.1016/j.prp.2024.155412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 06/07/2024] [Accepted: 06/13/2024] [Indexed: 06/20/2024]
Abstract
According to findings, long non-coding RNAs (lncRNAs) serves an integral part in growth and development of a variety of human malignancies, including Hepatoblastoma (HB). HB is a rare kind of carcinoma of the liver that mostly affects kids and babies under the age of three. Its manifestations include digestive swelling, abdominal discomfort, and losing weight. This thorough investigation digs into the many roles that lncRNAs serve in HB, giving views into their varied activities as well as possible therapeutic consequences. The function of lncRNAs in HB cell proliferation, apoptosis, migratory and penetrating capacities, epithelial-mesenchymal transition, and therapy tolerance is discussed. Various lncRNA regulatory roles are investigated in depth, yielding information on their effect on essential cell processes such as angiogenesis, apoptosis, immunity, and growth. Circulating lncRNAs are currently acknowledged as potential indications for the initial stages of identification of cancer, with the ability to diagnose as well as forecast. In addition to their diagnostic utility, lncRNAs provide curative opportunities as locations and actors, contributing to the expanding landscape of cancer research. Several HB-linked lncRNAs have been demonstrated to exhibit abnormal expression and are involved in tumor-like characteristics via DNA, RNA, or protein binding or encoding short peptides. As a result, a better knowledge of lncRNA instability might bring fresh perspectives into HB etiology as well as innovative strategies for HB early diagnosis and therapy. We describe the abnormalities of lncRNA expression in HB and their tumor-suppressive or carcinogenic activities during HB carcinogenesis in this study. Furthermore, we explore lncRNAs' diagnostic and therapeutic possibilities in HB.
Collapse
Affiliation(s)
- A Khuzaim Alzahrani
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Northern Border University, Arar 91431, Saudi Arabia
| | - Abida Khan
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| | - Neelam Singla
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur 302017, India
| | - Abdul Hai
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Northern Border University, Arar 91431, Saudi Arabia
| | - Abdullah R Alzahrani
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Al-Abidiyah, P.O. Box 13578, Makkah 21955, Saudi Arabia
| | - Mehnaz Kamal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | | | | | - Maitham Abdullah Al Hawaj
- Department of Pharmacy Practice, College of Clinical Pharmacy, King Faisal University, Al Ahsa 31982, Saudi Arabia
| | - Lulu Homeed Al Odaini
- Department of Ambulatory Care Pharmacy, King Fahad Medical City, Riyadh 12242, Saudi Arabia
| | - Tafadzwa Dzinamarira
- School of Health Systems and Public Health, University of Pretoria, Pretoria, South Africa
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia.
| |
Collapse
|
2
|
Zhang S, Jiang C, Jiang L, Chen H, Huang J, Gao X, Xia Z, Tran LJ, Zhang J, Chi H, Yang G, Tian G. Construction of a diagnostic model for hepatitis B-related hepatocellular carcinoma using machine learning and artificial neural networks and revealing the correlation by immunoassay. Tumour Virus Res 2023; 16:200271. [PMID: 37774952 PMCID: PMC10638043 DOI: 10.1016/j.tvr.2023.200271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 08/21/2023] [Accepted: 09/20/2023] [Indexed: 10/01/2023] Open
Abstract
HBV infection profoundly escalates hepatocellular carcinoma (HCC) susceptibility, responsible for a majority of HCC cases. HBV-driven immune-mediated hepatocyte impairment significantly fuels HCC progression. Regrettably, inconspicuous early HCC symptoms often culminate in belated diagnoses. Nevertheless, surgically treated early-stage HCC patients relish augmented five-year survival rates. In contrast, advanced HCC exhibits feeble responses to conventional interventions like radiotherapy, chemotherapy, and surgery, leading to diminished survival rates. This investigation endeavors to unearth diagnostic hallmark genes for HBV-HCC leveraging a bioinformatics framework, thus refining early HBV-HCC detection. Candidate genes were sieved via differential analysis and Weighted Gene Co-Expression Network Analysis (WGCNA). Employing three distinct machine learning algorithms unearthed three feature genes (HHIP, CXCL14, and CDHR2). Melding these genes yielded an innovative Artificial Neural Network (ANN) diagnostic blueprint, portending to alleviate patient encumbrance and elevate life quality. Immunoassay scrutiny unveiled accentuated immune damage in HBV-HCC patients relative to solitary HCC. Through consensus clustering, HBV-HCC was stratified into two subtypes (C1 and C2), the latter potentially indicating milder immune impairment. The diagnostic model grounded in these feature genes showcased robust and transferrable prognostic potentialities, introducing a novel outlook for early HBV-HCC diagnosis. This exhaustive immunological odyssey stands poised to expedite immunotherapeutic curatives' emergence for HBV-HCC.
Collapse
Affiliation(s)
- Shengke Zhang
- Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Chenglu Jiang
- Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Lai Jiang
- Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Haiqing Chen
- Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Jinbang Huang
- Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Xinrui Gao
- Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Zhijia Xia
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, 81377, Germany
| | - Lisa Jia Tran
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, 81377, Germany
| | - Jing Zhang
- Division of Basic Biomedical Sciences, The University of South Dakota Sanford School of Medicine, Vermillion, 57069, USA
| | - Hao Chi
- Clinical Medical College, Southwest Medical University, Luzhou, 646000, China.
| | - Guanhu Yang
- Department of Specialty Medicine, Ohio University, Athens, 45701, USA.
| | - Gang Tian
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
3
|
State of the art and perspectives in pediatric hepatocellular carcinoma. Biochem Pharmacol 2023; 207:115373. [PMID: 36513143 DOI: 10.1016/j.bcp.2022.115373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/29/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022]
Abstract
Hepatoblastoma (HB) and pediatric hepatocellular carcinoma (HCC) are rare primary malignant liver cancers in children and young adults. HB is the most common and accounts for about 70 % cases; it is usually diagnosed during the first 3 years of life. Instead, pediatric HCC is uncommon, and it is associated with a poor prognosis. Overall, the prognosis of pediatric HCC is dismal with 5-year event-free survival of <30 % as compared to >80 % for HB. Surgery approaches, either resection or transplant, remain the best chance for the cure of pediatric HCC. However, chemotherapy can be helpful as an adjuvant or neoadjuvant treatment. International groups have done trials in pediatric HCC with a chemotherapy regimen, based on cisplatin and doxorubicin (PLADO) as for HB, but the efficacy is limited. Sorafenib, a multi-kinase inhibitor, following positive results in adults and in a pilot study in children, is now tested in conjunction with chemotherapy in the PHITT phase III clinical trial. Some studies have been exploring the genetic profiles of patients to find biological hallmarks that determine the aggressiveness of pediatric HCC. Pathways involved in growth and differentiation are dysregulated and as demonstrated in HB and adult HCC, an important role of the Wnt/CTNNB1 pathway in the pathogenesis of pediatric HCC is also emerging. An extended molecular analysis of tumor samples could give information about pathways as possible targets of biological and immunotherapeutic agents bringing new pharmacological options for the treatment of pediatric HCC.
Collapse
|
4
|
Jiang J. Hedgehog signaling mechanism and role in cancer. Semin Cancer Biol 2022; 85:107-122. [PMID: 33836254 PMCID: PMC8492792 DOI: 10.1016/j.semcancer.2021.04.003] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/25/2021] [Accepted: 04/02/2021] [Indexed: 12/12/2022]
Abstract
Cell-cell communication through evolutionarily conserved signaling pathways governs embryonic development and adult tissue homeostasis. Deregulation of these signaling pathways has been implicated in a wide range of human diseases including cancer. One such pathway is the Hedgehog (Hh) pathway, which was originally discovered in Drosophila and later found to play a fundamental role in human development and diseases. Abnormal Hh pathway activation is a major driver of basal cell carcinomas (BCC) and medulloblastoma. Hh exerts it biological influence through a largely conserved signal transduction pathway from the activation of the GPCR family transmembrane protein Smoothened (Smo) to the conversion of latent Zn-finger transcription factors Gli/Ci proteins from their repressor (GliR/CiR) to activator (GliA/CiA) forms. Studies from model organisms and human patients have provided deep insight into the Hh signal transduction mechanisms, revealed roles of Hh signaling in a wide range of human cancers, and suggested multiple strategies for targeting this pathway in cancer treatment.
Collapse
Affiliation(s)
- Jin Jiang
- Department of Molecular Biology and Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390, USA.
| |
Collapse
|
5
|
Kolorz J, Demir S, Gottschlich A, Beirith I, Ilmer M, Lüthy D, Walz C, Dorostkar MM, Magg T, Hauck F, von Schweinitz D, Kobold S, Kappler R, Berger M. The Neurokinin-1 Receptor Is a Target in Pediatric Rhabdoid Tumors. Curr Oncol 2021; 29:94-110. [PMID: 35049682 PMCID: PMC8775224 DOI: 10.3390/curroncol29010008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/21/2021] [Accepted: 12/24/2021] [Indexed: 12/25/2022] Open
Abstract
Rhabdoid tumors (RT) are among the most aggressive tumors in early childhood. Overall survival remains poor, and treatment only effectively occurs at the cost of high toxicity and late adverse effects. It has been reported that the neurokinin-1 receptor/ substance P complex plays an important role in cancer and proved to be a promising target. However, its role in RT has not yet been described. This study aims to determine whether the neurokinin-1 receptor is expressed in RT and whether neurokinin-1 receptor (NK1R) antagonists can serve as a novel therapeutic approach in treating RTs. By in silico analysis using the cBio Cancer Genomics Portal we found that RTs highly express neurokinin-1 receptor. We confirmed these results by RT-PCR in both tumor cell lines and in human tissue samples of various affected organs. We demonstrated a growth inhibitory and apoptotic effect of aprepitant in viability assays and flow cytometry. Furthermore, this effect proved to remain when used in combination with the cytostatic cisplatin. Western blot analysis showed an upregulation of apoptotic signaling pathways in rhabdoid tumors when treated with aprepitant. Overall, our findings suggest that NK1R may be a promising target for the treatment of RT in combination with other anti-cancer therapies and can be targeted with the NK1R antagonist aprepitant.
Collapse
Affiliation(s)
- Julian Kolorz
- Research Laboratories, Department of Pediatric Surgery, Dr. von Hauner Children’s Hospital, Ludwig-Maximilians-University Munich, 80337 Munich, Germany; (J.K.); (S.D.); (D.L.); (D.v.S.); (R.K.)
| | - Salih Demir
- Research Laboratories, Department of Pediatric Surgery, Dr. von Hauner Children’s Hospital, Ludwig-Maximilians-University Munich, 80337 Munich, Germany; (J.K.); (S.D.); (D.L.); (D.v.S.); (R.K.)
| | - Adrian Gottschlich
- Center for Integrated Protein Science Munich (CIPSM) and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, 80337 Munich, Germany; (A.G.); (S.K.)
| | - Iris Beirith
- Department of General, Visceral, and Transplantation Surgery, University Hospital, Ludwig-Maximilians-University Munich, 81377 Munich, Germany; (I.B.); (M.I.)
| | - Matthias Ilmer
- Department of General, Visceral, and Transplantation Surgery, University Hospital, Ludwig-Maximilians-University Munich, 81377 Munich, Germany; (I.B.); (M.I.)
- German Center for Translational Cancer Research (DKTK), Partner Site Munich, 81377 Munich, Germany
| | - Daniel Lüthy
- Research Laboratories, Department of Pediatric Surgery, Dr. von Hauner Children’s Hospital, Ludwig-Maximilians-University Munich, 80337 Munich, Germany; (J.K.); (S.D.); (D.L.); (D.v.S.); (R.K.)
| | - Christoph Walz
- Institute of Pathology, Faculty of Medicine, Ludwig Maximilians-University Munich, 80337 Munich, Germany;
| | - Mario M. Dorostkar
- Center for Neuropathology, Ludwig-Maximilians-University Munich, 81377 Munich, Germany;
| | - Thomas Magg
- Department of Pediatrics, Dr. von Hauner Children’s Hospital, Ludwig-Maximilians-University Munich, 80337 Munich, Germany; (T.M.); (F.H.)
| | - Fabian Hauck
- Department of Pediatrics, Dr. von Hauner Children’s Hospital, Ludwig-Maximilians-University Munich, 80337 Munich, Germany; (T.M.); (F.H.)
| | - Dietrich von Schweinitz
- Research Laboratories, Department of Pediatric Surgery, Dr. von Hauner Children’s Hospital, Ludwig-Maximilians-University Munich, 80337 Munich, Germany; (J.K.); (S.D.); (D.L.); (D.v.S.); (R.K.)
| | - Sebastian Kobold
- Center for Integrated Protein Science Munich (CIPSM) and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, 80337 Munich, Germany; (A.G.); (S.K.)
- German Center for Translational Cancer Research (DKTK), Partner Site Munich, 81377 Munich, Germany
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), 85764 Neuherberg, Germany
| | - Roland Kappler
- Research Laboratories, Department of Pediatric Surgery, Dr. von Hauner Children’s Hospital, Ludwig-Maximilians-University Munich, 80337 Munich, Germany; (J.K.); (S.D.); (D.L.); (D.v.S.); (R.K.)
| | - Michael Berger
- Research Laboratories, Department of Pediatric Surgery, Dr. von Hauner Children’s Hospital, Ludwig-Maximilians-University Munich, 80337 Munich, Germany; (J.K.); (S.D.); (D.L.); (D.v.S.); (R.K.)
- Correspondence: ; Tel.: +49-89-4400-57859
| |
Collapse
|
6
|
The Role of the Hedgehog Pathway in Cholangiocarcinoma. Cancers (Basel) 2021; 13:cancers13194774. [PMID: 34638259 PMCID: PMC8507550 DOI: 10.3390/cancers13194774] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/17/2021] [Accepted: 09/21/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Cholangiocarcinoma (CCA) is one of the most refractory malignancies with a high mortality rate. Among all the pathways involved in CCA development, emerging evidence highlights Hedgehog (HH) signaling as a substantial player in CCA-genesis and development. The pro-tumoral function of HH provides potential therapeutic implications, and recently the use of HH inhibitors has paved the way for clinical application in various solid tumors. Targeting HH members, namely Hedgehog ligands, SMO transmembrane protein and GLI transcription factors may thus confer therapeutic options for the improvement of CCA treatment outcome. Abstract Cholangiocarcinoma (CCA) is a poorly treatable type of cancer and, along with hepatocellular carcinoma (HCC), is the predominant type of primitive liver cancer in adults. The lack of understanding of CCA biology has slowed down the identification of novel targets and the development of effective treatments. While tumors share some general characteristics, detailed knowledge of specific features is essential for the development of effectively tailored therapeutic approaches. The Hedgehog (HH) signaling cascade regulates stemness biology, embryonal development, tissue homeostasis, and cell proliferation and differentiation. Its aberrant activation has been associated with a variety of solid and hematological human malignancies. Several HH-inhibiting compounds have been indeed developed as potential anticancer agents in different types of tumors, with Smoothened and GLI inhibitors showing the most promising results. Beside its well-established function in other tumors, findings regarding the HH signaling in CCA are still controversial. Here we will give an overview of the most important clinical and molecular features of cholangiocarcinoma, and we will discuss the available evidence of the crosstalk between the HH signaling pathway and the cholangiocarcinoma cell biology.
Collapse
|
7
|
Lv D, Chen L, Du L, Zhou L, Tang H. Emerging Regulatory Mechanisms Involved in Liver Cancer Stem Cell Properties in Hepatocellular Carcinoma. Front Cell Dev Biol 2021; 9:691410. [PMID: 34368140 PMCID: PMC8339910 DOI: 10.3389/fcell.2021.691410] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 07/01/2021] [Indexed: 02/05/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the predominant form of primary liver cancer and one of the leading causes of cancer-related deaths worldwide. A growing body of evidence supports the hypothesis that HCC is driven by a population of cells called liver cancer stem cells (LCSCs). LCSCs have been proposed to contribute to malignant HCC progression, including promoting tumor occurrence and growth, mediating tumor metastasis, and treatment resistance, but the regulatory mechanism of LCSCs in HCC remains unclear. Understanding the signaling pathways responsible for LCSC maintenance and survival may provide opportunities to improve patient outcomes. Here, we review the current literature about the origin of LCSCs and the niche composition, describe the current evidence of signaling pathways that mediate LCSC stemness, then highlight several mechanisms that modulate LCSC properties in HCC progression, and finally, summarize the new developments in therapeutic strategies targeting LCSCs markers and regulatory pathways.
Collapse
Affiliation(s)
- Duoduo Lv
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Liyu Chen
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Lingyao Du
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Lingyun Zhou
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China.,State Key Laboratory of Biotherapy and Center of Infectious Diseases, Division of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
8
|
Li P, Zhang X, Murphy AJ, Costa M, Zhao X, Sun H. Downregulation of hedgehog-interacting protein (HHIP) contributes to hexavalent chromium-induced malignant transformation of human bronchial epithelial cells. Carcinogenesis 2021; 42:136-147. [PMID: 32710611 DOI: 10.1093/carcin/bgaa085] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/10/2020] [Accepted: 07/20/2020] [Indexed: 12/25/2022] Open
Abstract
Hexavalent chromium [Cr(VI)] is a potent human lung carcinogen. Multiple mechanisms have been proposed that contribute to Cr(VI)-induced lung carcinogenesis including oxidative stress, DNA damage, genomic instability and epigenetic modulation. However, the molecular mechanisms and pathways mediating Cr(VI) carcinogenicity have not been fully elucidated. Hedgehog (Hh) signaling is a key pathway that plays important roles in the formation of multiple tissues during embryogenesis and in the maintenance of stem cell populations in adults. Dysregulation of Hh signaling pathway has been reported in many human cancers. Here, we report a drastic reduction in both mRNA and protein levels of hedgehog-interacting protein (HHIP), a downstream target and a negative regulator of Hh signaling, in Cr(VI)-transformed cells. These findings point to a potential role of Hh signaling in Cr(VI)-induced malignant transformation and lung carcinogenesis. Cr(VI)-transformed cells exhibited DNA hypermethylation and silencing histone marks in the promoter region of HHIP, indicating that an epigenetic mechanism mediates Cr(VI)-induced silencing of HHIP. In addition, the major targets of Hh signaling (GLI1-3 and PTCH1) were significantly increased in Cr(VI)-transformed cells, suggesting an aberrant activation of Hh signaling in these cells. Moreover, ectopically expressing HHIP not only suppressed Hh signaling but also inhibited cell proliferation and anchorage-independent growth in Cr(VI)-transformed cells. In conclusion, these findings establish a novel regulatory mechanism underlying Cr(VI)-induced lung carcinogenesis and provide new insights for developing a better diagnostic and prognostic strategy for Cr(VI)-related human lung cancer.
Collapse
Affiliation(s)
- Peichao Li
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.,Department of Environmental Medicine, NYU School of Medicine, New York
| | - Xiaoru Zhang
- Department of Environmental Medicine, NYU School of Medicine, New York
| | - Anthony J Murphy
- Department of Environmental Medicine, NYU School of Medicine, New York
| | - Max Costa
- Department of Environmental Medicine, NYU School of Medicine, New York
| | - Xiaogang Zhao
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Hong Sun
- Department of Environmental Medicine, NYU School of Medicine, New York
| |
Collapse
|
9
|
Abstract
Hepatoblastoma (HB) is the predominant primary liver tumor in children. While the prognosis is favorable when the tumor can be resected, the outcome is dismal for patients with progressed HB. Therefore, a better understanding of the molecular mechanisms responsible for HB is imperative for early detection and effective treatment. Sequencing analysis of human HB specimens unraveled the pivotal role of Wnt/β-catenin pathway activation in this disease. Nonetheless, β-catenin activation alone does not suffice to induce HB, implying the need for additional alterations. Perturbations of several pathways, including Hippo, Hedgehog, NRF2/KEAP1, HGF/c-Met, NK-1R/SP, and PI3K/AKT/mTOR cascades and aberrant activation of c-MYC, n-MYC, and EZH2 proto-oncogenes, have been identified in HB, although their role requires additional investigation. Here, we summarize the current knowledge on HB molecular pathogenesis, the relevance of the preclinical findings for the human disease, and the innovative therapeutic strategies that could be beneficial for the treatment of HB patients.
Collapse
Affiliation(s)
- Yi Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China,Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California
| | - Antonio Solinas
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Stefano Cairo
- XenTech, Evry, France,Istituto di Ricerca Pediatrica, Padova, Italy
| | - Matthias Evert
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California
| | - Diego F. Calvisi
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| |
Collapse
|
10
|
Shen G, Shen H, Zhang J, Yan Q, Liu H. DNA methylation in Hepatoblastoma-a literature review. Ital J Pediatr 2020; 46:113. [PMID: 32758256 PMCID: PMC7409486 DOI: 10.1186/s13052-020-00877-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 07/29/2020] [Indexed: 12/19/2022] Open
Abstract
Hepatoblastoma (HB) is the most common malignant liver tumor in children. Abnormal activation of the Wnt/β-catenin signaling pathway plays an important role in the formation and development of HB. Genes in HB show a global hypomethylation change, accompanied by hypermethylation of specific tumor suppressor genes (TSGs). This article reviews the hypermethylation changes in several TSGs, such as RASSF1A, SOCS1, APC, HHIP, and P16, and analyzes the pathways and mechanisms of TSGs regulating gene expression. The role of the methylation-regulating enzymes DNA methyltransferases (DNMTs) and ten-eleven translocation (TET) family members enzymes in the methylation changes of HB was analyzed, and it was speculated that the occurrence of HB is partly due to the obstruction of liver differentiation in the early stage of differentiation. The origin cells may be incompletely differentiated hepatocytes remaining in the liver of children after birth. Therefore, further studying the role of methylation regulating enzymes in methylation changes in HB is a promising future research direction.
Collapse
Affiliation(s)
- Gang Shen
- Pediatric Surgery Department, Weifang Peoples' Hospital, Weifang, China
| | - Hongyu Shen
- Ultrasound Department, Weifang Haifushan Hospital, Weifang, China
| | - Jing Zhang
- Pediatric Surgery Department, Weifang Peoples' Hospital, Weifang, China
| | - Qingtao Yan
- Pediatric Surgery Department, Weifang Peoples' Hospital, Weifang, China
| | - Huixian Liu
- Dermatology Department, Weifang Peoples' Hospital, No. 151, Guangwen Street, Kuiwen District, Weifang, 261041, China.
| |
Collapse
|
11
|
Calvisi DF, Solinas A. Hepatoblastoma: current knowledge and promises from preclinical studies. Transl Gastroenterol Hepatol 2020; 5:42. [PMID: 32632393 DOI: 10.21037/tgh.2019.12.03] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 11/12/2019] [Indexed: 12/11/2022] Open
Abstract
The survival rate for patients with metastatic hepatoblastoma (HB) is steadily increased in the last thirty years from 27% to 79%. These achievements result from accurate risk stratification and effective chemotherapy and surgical care. However, patients with poor prognosis require more effective therapies. Recent years have witnessed new insights on the biology of HB, setting the stage for molecular classification and new targets of therapy. We review here the molecular pathology of HB, focusing on the driver genes involved in the process of oncogenesis and the identification of novel targets. We also address the role of in vivo models in elucidating the mechanisms of development of this disease and the pre-clinical phase of new treatment modalities.
Collapse
Affiliation(s)
- Diego F Calvisi
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Antonio Solinas
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| |
Collapse
|
12
|
Liver regeneration and liver metastasis. Semin Cancer Biol 2020; 71:86-97. [PMID: 32532594 DOI: 10.1016/j.semcancer.2020.05.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/18/2020] [Accepted: 05/18/2020] [Indexed: 12/12/2022]
Abstract
Surgical resection for primary and secondary hepatic neoplasms provides the best chance of cure. Advanced surgical techniques such as portal vein embolisation, two-staged hepatectomy and associated liver partition and portal vein ligation for staged-hepatectomy (ALPPS) have facilitated hepatic resection in patients with previously unresectable, bi-lobar disease. These techniques are frequently employed to ensure favourable clinical outcomes and avoid potentially fatal post-operative complications such as small for size syndrome and post-hepatectomy liver failure. However, they rely on the innate ability of the liver to regenerate. As our knowledge of liver organogenesis, liver regeneration and hepatocarcinogenesis has expanded in recent decades it has come to light that liver regeneration may also drive tumour recurrence. Clinical studies in patients undergoing portal vein embolisation indicate that tumours may progress following the procedure in concordance with liver regeneration and hypertrophy, however overall survival in these patients has not been shown to be worse. In this article, we delve into the mechanisms underlying liver regeneration to better understand the complex ways in which this may affect tumour behaviour and ultimately inform clinical decisions.
Collapse
|
13
|
Wang Z, Song L, Ye Y, Li W. Long Noncoding RNA DIO3OS Hinders Cell Malignant Behaviors of Hepatocellular Carcinoma Cells Through the microRNA-328/Hhip Axis. Cancer Manag Res 2020; 12:3903-3914. [PMID: 32547226 PMCID: PMC7259459 DOI: 10.2147/cmar.s245990] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 04/30/2020] [Indexed: 12/24/2022] Open
Abstract
Background The decline of a long non-coding RNA (lncRNA) DIO3OS was implicated in a plethora of cancers, while the relevance in hepatocellular carcinoma (HCC) has not been mentioned. Accordingly, we set to determine the functional role of DIO3OS and the molecular mechanism in HCC progression. Materials and Methods The differentially expressed lncRNAs, mRNAs, and microRNAs (miRNAs) were obtained through the datasets GSE101728 and GES57555. Afterwards, DIO3OS was enhanced in HCC cells to examine the behavior changes. Subcellular localization of DIO3OS was determined through website prediction and experimental validation. The expression of Hedgehog (Hh) signaling pathway-related genes was detected. The effects of DIO3OS overexpression on tumor growth were evaluated as well. Results DIO3OS was lower in HCC tissues and cells, while upregulation of DIO3OS repressed malignant biological behavior both in vitro and in vivo. DIO3OS, localized in the cytoplasm, inhibited the occurrence of HCC by disrupting the Hh pathway by sponging miR-328 to mediate Hh interacting protein (Hhip). Conclusion All in all, the obtained data suggested that DIO3OS interacted with Hhip-dependent Hh signaling pathway to inhibit HCC progression through binding to miR-328, which may be a potent therapeutic target for HCC.
Collapse
Affiliation(s)
- Zhanpeng Wang
- Department of Hepatobiliary-Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Changchun 130000, People's Republic of China
| | - Lina Song
- Department of Laboratory Medicine Center, China-Japan Union Hospital of Jilin University, Changchun 130000, People's Republic of China
| | - Yanshuo Ye
- Department of Hepatobiliary-Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Changchun 130000, People's Republic of China
| | - Wei Li
- Department of Hepatobiliary-Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Changchun 130000, People's Republic of China
| |
Collapse
|
14
|
Downregulation of SFRP1 is a protumorigenic event in hepatoblastoma and correlates with beta-catenin mutations. J Cancer Res Clin Oncol 2020; 146:1153-1167. [PMID: 32189106 PMCID: PMC7142044 DOI: 10.1007/s00432-020-03182-1] [Citation(s) in RCA: 145] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 03/11/2020] [Indexed: 12/20/2022]
Abstract
Background Hepatoblastoma (HB) and pediatric hepatocellular carcinoma (HCC) are the most common malignant liver tumors in childhood. Both tumor types exhibit genetic and epigenetic alterations in the WNT/β-catenin signaling pathway, which is a key regulator of liver progenitor cells in embryonic development. The tumors demonstrate a high rate of β-catenin mutations and gene expression changes of several WNT antagonists. However, the role of the WNT inhibitory factor secreted frizzled-related protein 1 (SFRP1) has not been addressed in pediatric liver cancer so far. Results In our study, we investigated the gene expression level, DNA methylation status and functional relevance of SFRP1 in HB cell lines and in pediatric liver tumor patient samples. SFRP1 was downregulated due to DNA promoter methylation in all tested HB cell lines. Overexpression of SFRP1 in HB cell lines diminished tumor cell proliferation, colony formation and migration potential. In addition, the SFRP1-expressing HB cell lines showed reduced WNT/β-catenin signaling pathway activity and decreased expression of WNT target genes. To evaluate the utility of SFRP1 as a biomarker in pediatric liver cancer, we determined the gene expression level and DNA methylation status of SFRP1 in 45 pediatric liver tumor patient samples. The correlation analysis of different clinical parameters and tumor characteristics revealed a significant correlation of reduced SFRP1 expression with the presence of mutant β-catenin. The methylation status of SFRP1 was furthermore associated to a pediatric liver tumor type with HCC-like characteristics, TERT mutations and an older age at diagnosis. Conclusion Altogether, our data demonstrate that the epigenetic suppression of the WNT/β-catenin antagonist SFRP1 has an important impact on the malignant behavior of HB cells. Although SFRP1 methylation is a common event in HCC-like pediatric liver tumors, its potential as a prognostic or diagnostic biomarker needs to be further investigated. Electronic supplementary material The online version of this article (10.1007/s00432-020-03182-1) contains supplementary material, which is available to authorized users.
Collapse
|
15
|
Choo Z, Loh AHP, Chen ZX. Destined to Die: Apoptosis and Pediatric Cancers. Cancers (Basel) 2019; 11:cancers11111623. [PMID: 31652776 PMCID: PMC6893512 DOI: 10.3390/cancers11111623] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/20/2019] [Accepted: 10/22/2019] [Indexed: 01/10/2023] Open
Abstract
Apoptosis (programmed cell death) is a systematic and coordinated cellular process that occurs in physiological and pathophysiological conditions. Sidestepping or resisting apoptosis is a distinct characteristic of human cancers including childhood malignancies. This review dissects the apoptosis pathways implicated in pediatric tumors. Understanding these pathways not only unraveled key molecules that may serve as potential targets for drug discovery, but also molecular nodes that integrate with other signaling networks involved in processes such as development. This review presents current knowledge of the complex regulatory system that governs apoptosis with respect to other processes in pediatric cancers, so that fresh insights may be derived regarding treatment resistance or for more effective treatment options.
Collapse
Affiliation(s)
- Zhang'e Choo
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore.
| | - Amos Hong Pheng Loh
- VIVA-KKH Pediatric Brain and Solid Tumor Program, KK Women's and Children's Hospital, Singapore 229899, Singapore.
- Department of Pediatric Surgery, KK Women's and Children's Hospital, Singapore 229899, Singapore.
| | - Zhi Xiong Chen
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore.
- VIVA-KKH Pediatric Brain and Solid Tumor Program, KK Women's and Children's Hospital, Singapore 229899, Singapore.
- National University Cancer Institute, Singapore, Singapore 119074, Singapore.
| |
Collapse
|
16
|
Eberherr C, Beck A, Vokuhl C, Becker K, Häberle B, Von Schweinitz D, Kappler R. Targeting excessive MYCN expression using MLN8237 and JQ1 impairs the growth of hepatoblastoma cells. Int J Oncol 2019; 54:1853-1863. [PMID: 30864675 DOI: 10.3892/ijo.2019.4741] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 01/23/2019] [Indexed: 11/06/2022] Open
Abstract
Hepatoblastoma (HB) is the most common liver tumor in children under the age of 3 years worldwide. While many patients achieve good outcomes with surgical resection and conventional chemotherapy, there is still a high‑risk population that exhibits a poor treatment response and unfavorable prognosis, which warrants the search for novel treatment options. In recent years, it has become clear that genetic events alone are not sufficient to explain the aggressive phenotype of this embryonal malignancy. Instead, epigenetic modifications and aberrant gene expression seem to be key drivers of HB. In the present study, expression analyses such as reverse transcription‑quantitative polymerase chain reaction revealed that the oncogene, MYCN proto‑oncogene basic‑helix‑loop‑helix transcription factor (MYCN) was upregulated in HB and other pediatric liver tumors, due to the transcriptional activity of its antisense transcript MYCN opposite strand (MYCNOS). Pyrosequencing demonstrated the hypomethylated regions in the promoter of MYCN and MYCNOS, suggesting that an epigenetic mechanism may underlie the induction of aberrant expression. Transient MYCN knockdown in HB cells resulted in growth inhibition over time. In addition, treating HB cells with the MYCN inhibitors JQ1 and MLN8237 led to the significant downregulation of MYCN either at the mRNA or protein levels, respectively. The underlying mechanism of action of the two inhibitors was revealed to be associated with the induction of dose‑dependent growth arrest, by arresting cells at either the G1/G0 or G2 phase. Furthermore, MLN8237 and JQ1 were able to cause spindle disturbances and/or apoptosis in HB cells. The present results suggest that MYCN may be a promising biomarker for HB and a potential therapeutic target in patients with tumors overexpressing MYCN.
Collapse
Affiliation(s)
- Corinna Eberherr
- Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, University Hospital, LMU Munich, D‑80337 Munich, Germany
| | - Alexander Beck
- Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, University Hospital, LMU Munich, D‑80337 Munich, Germany
| | - Christian Vokuhl
- Institute of Paidopathology, Pediatric Tumor Registry, Christian‑Albrecht's‑University Kiel, D‑24105 Kiel, Germany
| | - Kristina Becker
- Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, University Hospital, LMU Munich, D‑80337 Munich, Germany
| | - Beate Häberle
- Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, University Hospital, LMU Munich, D‑80337 Munich, Germany
| | - Dietrich Von Schweinitz
- Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, University Hospital, LMU Munich, D‑80337 Munich, Germany
| | - Roland Kappler
- Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, University Hospital, LMU Munich, D‑80337 Munich, Germany
| |
Collapse
|
17
|
High Risk of Hepatocellular Carcinoma Development in Fibrotic Liver: Role of the Hippo-YAP/TAZ Signaling Pathway. Int J Mol Sci 2019; 20:ijms20030581. [PMID: 30700007 PMCID: PMC6387126 DOI: 10.3390/ijms20030581] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 01/24/2019] [Accepted: 01/28/2019] [Indexed: 02/07/2023] Open
Abstract
Liver cancer is the fourth leading cause of cancer-related death globally, accounting for approximately 800,000 deaths annually. Hepatocellular carcinoma (HCC) is the most common type of liver cancer, making up about 80% of cases. Liver fibrosis and its end-stage disease, cirrhosis, are major risk factors for HCC. A fibrotic liver typically shows persistent hepatocyte death and compensatory regeneration, chronic inflammation, and an increase in reactive oxygen species, which collaboratively create a tumor-promoting microenvironment via inducing genetic alterations and chromosomal instability, and activating various oncogenic molecular signaling pathways. In this article, we review recent advances in fields of liver fibrosis and carcinogenesis, and consider several molecular signaling pathways that promote hepato-carcinogenesis under the microenvironment of liver fibrosis. In particular, we pay attention to emerging roles of the Hippo-YAP/TAZ signaling pathway in stromal activation, hepatic fibrosis, and liver cancer.
Collapse
|
18
|
Sun H, Ni SJ, Ye M, Weng W, Zhang Q, Zhang M, Tan C, Wang L, Huang D, Du X, Xu M, Sheng W. Hedgehog Interacting Protein 1 is a Prognostic Marker and Suppresses Cell Metastasis in Gastric Cancer. J Cancer 2018; 9:4642-4649. [PMID: 30588248 PMCID: PMC6299386 DOI: 10.7150/jca.27686] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 09/14/2018] [Indexed: 01/22/2023] Open
Abstract
Background: The gene Hedgehog interacting protein (HHIP) is a pivotal morphogen for multiple developmental processes. However, the expression and clinical correlation of HHIP in gastric cancer (GC) has not been fully investigated. Here, we aimed to explore the expression of HHIP in gastric cancer (GC) and evaluate its clinicopathological and functional correlations. Methods: The expression of HHIP mRNA was first determined in the Human Protein Atlas (HPA) and The Cancer Genome Atlas (TCGA) GC database and then validated by RT-qPCR (n = 41) and immunohistochemistry (IHC, n = 95) in a cohort of in-house GC patients and in 29 cases of gastric intraepithelial neoplasia (GIN). The clinicopathological and functional relationship of HHIP with GC were also analyzed. Results: We found that HHIP mRNA were significantly downregulated in GC in the TCGA and HPA databases, as well as in our in-house cohort (P < 0.05). HHIP mRNA is mainly located in the cell nucleus, while HHIP protein is mainly located in the cell cytoplasm. Moreover, the HHIP protein level in the GIN tissues was significantly higher than that in the GC tissues (P < 0.001) and significantly lower than that in adjacent normal controls (P < 0.001). In addition, low HHIP expression was correlated with lymphatic metastasis (P = 0.041), pTNM stage (P = 0.007) and nervous system invasion (P = 0.001). Furthermore, we observed strong positive correlations between HHIP protein expression and overall survival (P < 0.001) and disease-free survival (P = 0.027) in GC patients. HHIP protein expression was an independent prognostic factor for overall survival (P < 0.001). Functional experimental results showed that overexpression of HHIP attenuated the migration and invasion ability of GC cells (P < 0.01). Conclusion: HHIP may be a promising tumor metastatic-suppressor and prognostic biomarker for gastric cancer.
Collapse
Affiliation(s)
- Hui Sun
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Shu Juan Ni
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Min Ye
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Weiwei Weng
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Qiongyan Zhang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Meng Zhang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Cong Tan
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Lei Wang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Dan Huang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Xiang Du
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Midie Xu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Weiqi Sheng
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Institute of Pathology, Fudan University, Shanghai 200032, China
| |
Collapse
|
19
|
Virzì A, Roca Suarez AA, Baumert TF, Lupberger J. Oncogenic Signaling Induced by HCV Infection. Viruses 2018; 10:v10100538. [PMID: 30279347 PMCID: PMC6212953 DOI: 10.3390/v10100538] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 09/29/2018] [Accepted: 09/30/2018] [Indexed: 02/07/2023] Open
Abstract
The liver is frequently exposed to toxins, metabolites, and oxidative stress, which can challenge organ function and genomic stability. Liver regeneration is therefore a highly regulated process involving several sequential signaling events. It is thus not surprising that individual oncogenic mutations in hepatocytes do not necessarily lead to cancer and that the genetic profiles of hepatocellular carcinomas (HCCs) are highly heterogeneous. Long-term infection with hepatitis C virus (HCV) creates an oncogenic environment by a combination of viral protein expression, persistent liver inflammation, oxidative stress, and chronically deregulated signaling events that cumulate as a tipping point for genetic stability. Although novel direct-acting antivirals (DAA)-based treatments efficiently eradicate HCV, the associated HCC risk cannot be fully eliminated by viral cure in patients with advanced liver disease. This suggests that HCV may persistently deregulate signaling pathways beyond viral cure and thereby continue to perturb cancer-relevant gene function. In this review, we summarize the current knowledge about oncogenic signaling pathways derailed by chronic HCV infection. This will not only help to understand the mechanisms of hepatocarcinogenesis but will also highlight potential chemopreventive strategies to help patients with a high-risk profile of developing HCC.
Collapse
Affiliation(s)
- Alessia Virzì
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 67000 Strasbourg, France.
- Université de Strasbourg, 67000 Strasbourg, France.
| | - Armando Andres Roca Suarez
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 67000 Strasbourg, France.
- Université de Strasbourg, 67000 Strasbourg, France.
| | - Thomas F Baumert
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 67000 Strasbourg, France.
- Université de Strasbourg, 67000 Strasbourg, France.
- Pôle Hépato-digestif, Institut Hospitalo-universitaire, Hôpitaux Universitaires de Strasbourg, 67000 Strasbourg, France.
| | - Joachim Lupberger
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 67000 Strasbourg, France.
- Université de Strasbourg, 67000 Strasbourg, France.
| |
Collapse
|
20
|
Beck A, Trippel F, Wagner A, Joppien S, Felle M, Vokuhl C, Schwarzmayr T, Strom TM, von Schweinitz D, Längst G, Kappler R. Overexpression of UHRF1 promotes silencing of tumor suppressor genes and predicts outcome in hepatoblastoma. Clin Epigenetics 2018; 10:27. [PMID: 29507645 PMCID: PMC5833129 DOI: 10.1186/s13148-018-0462-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 02/21/2018] [Indexed: 02/02/2023] Open
Abstract
Background Hepatoblastoma (HB) is the most common liver tumor of childhood and occurs predominantly within the first 3 years of life. In accordance to its early manifestation, HB has been described to display an extremely low mutation rate. As substitute, epigenetic modifiers seem to play an exceptional role in its tumorigenesis, which holds promise to develop targeted therapies and establish biomarkers for patient risk stratification. Results We examined the role of a newly described protein complex consisting of three epigenetic regulators, namely E3 ubiquitin-like containing PHD and RING finger domain 1 (UHRF1), ubiquitin-specific-processing protease 7 (USP7), and DNA methyltransferase 1 (DNMT1), in HB. We found the complex to be located on the promoter regions of the pivotal HB-associated tumor suppressor genes (TSGs) HHIP, IGFBP3, and SFRP1 in HB cells, thereby leading to strong repression through DNA methylation and histone modifications. Consequently, knockdown of UHRF1 led to DNA demethylation and loss of the repressive H3K9me2 histone mark at the TSG loci with their subsequent transcriptional reactivation. The observed growth impairment of HB cells upon UHRF1 knockdown could be attributed to reduced expression of genes involved in cell cycle progression, negative regulation of cell death, LIN28B signaling, and the adverse 16-gene signature, as revealed by global RNA sequencing. Clinically, overexpression of UHRF1 in primary tumor tissues was significantly associated with poor survival and the prognostic high-risk 16-gene signature. Conclusion These findings suggest that UHRF1 is critical for aberrant TSG silencing and sustained growth signaling in HB and that UHRF1 overexpression levels might serve as a prognostic biomarker and potential molecular target for HB patients. Electronic supplementary material The online version of this article (10.1186/s13148-018-0462-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alexander Beck
- 1Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University Munich, Lindwurmstr. 2a, 80337 Munich, Germany
| | - Franziska Trippel
- 1Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University Munich, Lindwurmstr. 2a, 80337 Munich, Germany
| | - Alexandra Wagner
- 1Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University Munich, Lindwurmstr. 2a, 80337 Munich, Germany
| | - Saskia Joppien
- 1Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University Munich, Lindwurmstr. 2a, 80337 Munich, Germany
| | - Max Felle
- 2Department of Biochemistry III, University Regensburg, Regensburg, Germany
| | - Christian Vokuhl
- 3Institute of Paidopathology, Pediatric Tumor Registry, Christian-Albrecht's-University Kiel, Kiel, Germany
| | - Thomas Schwarzmayr
- 4Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany.,5Institute of Human Genetics, Technische Universität München, Munich, Germany
| | - Tim M Strom
- 4Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany.,5Institute of Human Genetics, Technische Universität München, Munich, Germany
| | - Dietrich von Schweinitz
- 1Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University Munich, Lindwurmstr. 2a, 80337 Munich, Germany
| | - Gernot Längst
- 2Department of Biochemistry III, University Regensburg, Regensburg, Germany
| | - Roland Kappler
- 1Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University Munich, Lindwurmstr. 2a, 80337 Munich, Germany
| |
Collapse
|
21
|
Tracking cellular and molecular changes in a species-specific manner during experimental tumor progression in vivo. Oncotarget 2018; 9:16149-16162. [PMID: 29662633 PMCID: PMC5882324 DOI: 10.18632/oncotarget.24598] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 02/21/2018] [Indexed: 01/29/2023] Open
Abstract
Hepatoblastoma (HBL) is a pediatric liver cancer with defined molecular alterations driving its progression. Here, we describe an animal model for HBL on the chick chorioallantoic membrane (CAM), which recapitulates relevant features of HBL in patients. Expression of classic tumor-associated proteins such as β-catenin, EpCAM and CK19 was maintained in acini-like organized tumors on CAM, as was synthesis of AFP, a tumor marker used for monitoring patient response. RNA sequencing revealed an unexpected molecular evolution of HBL cells on the CAM, with significant deregulation of more than 6,000 genes including more than half of all HOX genes. Bioinformatic analysis distinguish between tumor cell-expressed genes and chick genes, thereby shedding new light on the complex interactions taking place during HBL progression. Importantly, human tumor suppressive ribosomal genes were downregulated after implantation, whereas mitochondrial genes encoding for anti-apoptotic peptides were strongly induced in vivo. Meprin-1α expression was increased during evolution of CAM tumors and confirmed by immunohistochemistry. Cisplatin, a commonly used chemotherapeutic agent for HBL, showed significant anti-tumoral effects. Our results broaden the understanding of the molecular adaptation process of human cancer cells to the microenvironment and might help to elaborate novel therapeutic concepts for the treatment of this pediatric liver tumor.
Collapse
|
22
|
Machado MV, Diehl AM. Hedgehog signalling in liver pathophysiology. J Hepatol 2018; 68:550-562. [PMID: 29107151 PMCID: PMC5957514 DOI: 10.1016/j.jhep.2017.10.017] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 10/11/2017] [Accepted: 10/18/2017] [Indexed: 12/13/2022]
Abstract
Liver disease remains a leading cause of mortality worldwide despite recent successes in the field of viral hepatitis, because increases in alcohol consumption and obesity are fuelling an epidemic of chronic fatty liver disease for which there are currently no effective medical therapies. About 20% of individuals with chronic liver injury ultimately develop end-stage liver disease due to cirrhosis. Hence, treatments to prevent and reverse cirrhosis in individuals with ongoing liver injury are desperately needed. The development of successful treatments requires an improved understanding of the mechanisms controlling liver disease progression. The liver responds to diverse insults with a conserved wound healing response, suggesting that it might be generally beneficial to optimise pathways that are crucial for effective liver repair. The Hedgehog pathway has emerged as a potential target based on compelling preclinical and clinical data, which demonstrate that it critically regulates the liver's response to injury. Herein, we will summarise evidence of the Hedgehog pathway's role in liver disease and discuss how modulating pathway activity might be applied to improve liver disease outcomes.
Collapse
Affiliation(s)
- Mariana Verdelho Machado
- Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA,Gastroenterology Department, Hospital de Santa Maria, CHLN, Lisbon, Portugal
| | - Anna Mae Diehl
- Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
23
|
Gao L, Zhang Z, Zhang P, Yu M, Yang T. Role of canonical Hedgehog signaling pathway in liver. Int J Biol Sci 2018; 14:1636-1644. [PMID: 30416378 PMCID: PMC6216024 DOI: 10.7150/ijbs.28089] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Accepted: 08/01/2018] [Indexed: 12/19/2022] Open
Abstract
Hedgehog (Hh) signaling pathway plays an important role in embryonic development. It becomes reactivated in many types of acute and chronic liver injuries. Hh signaling is required for liver regeneration, regulates capillarisation, controls the fates of hepatic stellate cells, promotes liver fibrosis and liver cancers. In this review, we summarize the current knowledge of the role of canonical Hh signaling pathway in adult liver. This help to understand the pathogenesis of liver diseases and find out the new effective targeted therapeutic strategies for liver diseases treatments.
Collapse
Affiliation(s)
- Lili Gao
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Zhenya Zhang
- Department of general surgery, Hebei Medical University Fourth Hospital, Shijiazhuang, 050011, China
| | - Peng Zhang
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Minghua Yu
- Department of Oncology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
- ✉ Corresponding authors: Dr. Minghua Yu, Department of Oncology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China. Phone: 86-21-68030812; E-mail: and Dr. Tao Yang, Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Shanghai 201399, China. Phone: 86-21-68036516; E-mail:
| | - Tao Yang
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
- ✉ Corresponding authors: Dr. Minghua Yu, Department of Oncology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China. Phone: 86-21-68030812; E-mail: and Dr. Tao Yang, Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Shanghai 201399, China. Phone: 86-21-68036516; E-mail:
| |
Collapse
|
24
|
Affiliation(s)
- Matthias Ilmer
- Department of General, Visceral, and Transplantation Surgery, Hospital of the LMU Munich, Munich, Germany
| | - Michael Berger
- Department of Pediatric Surgery, Research Laboratories, Dr. von Hauner Children's Hospital, Ludwig Maximilians University Munich, Munich, Germany.,Department of Surgery, Emory Transplant Center, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
25
|
Stem cell, biomaterials and growth factors therapy for hepatocellular carcinoma. Biomed Pharmacother 2017; 88:1046-1053. [PMID: 28192881 DOI: 10.1016/j.biopha.2017.01.154] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 01/26/2017] [Accepted: 01/26/2017] [Indexed: 12/14/2022] Open
Abstract
Hepatocellular carcinoma is an antecedent of liver illnesses, including viral hepatitis, alcohol abuse, or metabolic disease. Transforming growth factor-Beta (TGF-b) plays an important role in creating a favorable microenvironment for tumor cell growth via two major mechanisms: an intrinsic activity as an autocrine growth factor and an extrinsic activity by inducing microenvironment changes. Recently stem cell therapy as also been a promising and potential treatment for liver cancer and in addition signaling pathways like GF/GFR systems, SDF-1α/CXC4 ligand receptor interaction and PI3K/Akt signaling, and cytokines has been identified to regulate cell fate decisions, and can be utilized to positively influence cell therapy outcomes. Thus stem cell-based therapy, together with signaling pathways can become a practical option in regenerative processes for replacing dead hepatocytes cells. Targeted drug delivery systems (TDDS) via biomaterials are presently been explored for cancer therapeutics especially liver cancer as it allows the enhancement of drug concentration in the liver and decrease the dosage and side effects. This review is intended to give a comprehensive summary of available liver cancer therapy using stem cells, growth factor and biomaterials.
Collapse
|
26
|
Abstract
Hepatoblastoma is the most common primary malignant hepatic tumor of infancy and childhood, occurring predominantly in the first two years of life. The management of hepatoblastoma has changed markedly over the last 3 decades; neoadjuvant chemotherapy is now standard, particularly in unresectable tumors resulting in considerable preoperative tumor shrinkage and sometimes near total ablation of the tumor. A 20 month old infant was incidentally found to have a 7.6cm right sided retroperitoneal tumor on routine screening ultrasonography for left ureteral stenosis. Serum alpha fetoprotein was elevated. Biopsy revealed hepatoblastoma, mixed epithelial and embryonal type without mesenchymal elements. He underwent neoadjuvant chemotherapy. Although the tumor had decreased considerably in size, close proximity to major vascular structures precluded safe resection. Liver transplantation was performed; the explanted liver showed complete tumor necrosis with no residual malignancy. The postoperative course was uncomplicated and he is continuing on sixth cycle of chemotherapy.
Collapse
Affiliation(s)
- Divya Sharma
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, United States
| | - Girish Subbarao
- Department of Pediatric Gastroenterology, Hepatology and Nutrition, Indiana University School of Medicine, United States
| | - Romil Saxena
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, United States.
| |
Collapse
|
27
|
Beck A, Eberherr C, Hagemann M, Cairo S, Häberle B, Vokuhl C, von Schweinitz D, Kappler R. Connectivity map identifies HDAC inhibition as a treatment option of high-risk hepatoblastoma. Cancer Biol Ther 2016; 17:1168-1176. [PMID: 27635950 DOI: 10.1080/15384047.2016.1235664] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Hepatoblastoma (HB) is the most common liver tumor of childhood, usually occurring in children under the age of 3 y. The prognosis of patients presenting with distant metastasis, vascular invasion and advanced tumor stages remains poor and children that do survive often face severe late effects from the aggressive chemotherapy regimen. To identify potential new therapeutics for high risk HB we used a 1,000-gene expression signature as input for a Connectivity Map (CMap) analysis, which predicted histone deacetylase (HDAC) inhibitors as a promising therapy option. Subsequent expression analysis of primary HB and HB cell lines revealed a general overexpression of HDAC1 and HDAC2, which has been suggested to be predictive for the efficacy of HDAC inhibition. Accordingly, treatment of HB cells with the HDAC inhibitors SAHA and MC1568 resulted in a potent reduction of cell viability, induction of apoptosis, reactivation of epigenetically suppressed tumor suppressor genes, and the reversion of the 16-gene HB classifier toward the more favorable expression signature. Most importantly, the combination of HDAC inhibitors and cisplatin - a major chemotherapeutic agent of HB treatment - revealed a strong synergistic effect, even at significantly reduced doses of cisplatin. Our findings suggest that HDAC inhibitors skew HB cells toward a more favorable prognostic phenotype through changes in gene expression, thus indicating a targeted molecular mechanism that seems to enhance the anti-proliferative effects of conventional chemotherapy. Thus, adding HDAC inhibitors to the treatment regimen of high risk HB could potentially improve outcomes and reduce severe late effects.
Collapse
Affiliation(s)
- Alexander Beck
- a Department of Pediatric Surgery, Dr. von Hauner Children's Hospital , Ludwig-Maximilians-University Munich , Munich , Germany
| | - Corinna Eberherr
- a Department of Pediatric Surgery, Dr. von Hauner Children's Hospital , Ludwig-Maximilians-University Munich , Munich , Germany
| | - Michaela Hagemann
- a Department of Pediatric Surgery, Dr. von Hauner Children's Hospital , Ludwig-Maximilians-University Munich , Munich , Germany
| | - Stefano Cairo
- b XenTech , 4 rue Pierre Fontaine , Evry , France.,c University of Ferrara, LTTA Center, Department of Morphology , Surgery and Experimental Medicine, Via Fossato di Mortara , Ferrara , Italy
| | - Beate Häberle
- a Department of Pediatric Surgery, Dr. von Hauner Children's Hospital , Ludwig-Maximilians-University Munich , Munich , Germany
| | - Christian Vokuhl
- d Institute of Paidopathology, Pediatric Tumor Registry, Christian-Albrechts-University Kiel , Kiel , Germany
| | - Dietrich von Schweinitz
- a Department of Pediatric Surgery, Dr. von Hauner Children's Hospital , Ludwig-Maximilians-University Munich , Munich , Germany
| | - Roland Kappler
- a Department of Pediatric Surgery, Dr. von Hauner Children's Hospital , Ludwig-Maximilians-University Munich , Munich , Germany
| |
Collapse
|
28
|
Paluszczak J, Wiśniewska D, Kostrzewska-Poczekaj M, Kiwerska K, Grénman R, Mielcarek-Kuchta D, Jarmuż-Szymczak M. Prognostic significance of the methylation of Wnt pathway antagonists-CXXC4, DACT2, and the inhibitors of sonic hedgehog signaling-ZIC1, ZIC4, and HHIP in head and neck squamous cell carcinomas. Clin Oral Investig 2016; 21:1777-1788. [PMID: 27553089 PMCID: PMC5442212 DOI: 10.1007/s00784-016-1946-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 08/16/2016] [Indexed: 12/18/2022]
Abstract
Objectives Aberrations in Wnt and Shh signaling pathways are related to the pathogenesis of head and neck carcinomas, and their activation frequently results from epigenetic alterations. This study aimed to assess the frequency of methylation of negative regulators of Wnt signaling: CXXC4, DACT2, HDPR1, and FBXW11 and Shh signaling: HHIP, PTCH1, SUFU, ZIC1, and ZIC4 and correlate it with clinicopathological features in this group of patients. Materials and methods Methylation-specific PCR was used to detect gene promoter methylation, and real-time PCR was used to assess gene expression level. Results The analysis of the occurrence of gene promoter methylation in head and neck carcinoma cell lines indicated that CXXC4, DACT2, HHIP, ZIC1, and ZIC4 are methylated in these tumors. These genes were further analyzed in tumor sections from oral and laryngeal cancer patients. Gene methylation rate was higher in laryngeal tumors. The methylation index in tumor samples correlated with the overall survival in a subgroup of oral cancer patients who died of the disease. Moreover, ZIC4 methylation correlated with lymph node involvement in oral cancer patients. Conclusions Our findings corroborate that the activation of Wnt signaling in head and neck squamous cell carcinoma (HNSCC) is related to epigenetic silencing of its negative regulators. Moreover, the results indicate that the same mechanism of activation may operate in the case of Shh signaling. Clinical relevance The methylation of ZIC4 may be considered a new prognostic marker in oral cavity and oropharyngeal tumors. Further investigations should determine the diagnostic significance of methylation of ZIC4, HHIP, and DACT2 in head and neck carcinomas.
Collapse
Affiliation(s)
- Jarosław Paluszczak
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, ul. Święcickiego 4, 60-781, Poznań, Poland.
| | - Dorota Wiśniewska
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, ul. Święcickiego 4, 60-781, Poznań, Poland
| | | | - Katarzyna Kiwerska
- Department of Cancer Genetics, Institute of Human Genetics, Polish Academy of Sciences, Poznań, Poland
| | - Reidar Grénman
- Department of Otorhinolaryngology, Head and Neck Surgery and Department of Medical Biochemistry, Turku University Central Hospital and Turku University, Turku, Finland
| | - Daniela Mielcarek-Kuchta
- Department of Otolaryngology and Laryngological Oncology, Poznan University of Medical Sciences, Poznań, Poland
| | | |
Collapse
|
29
|
Lin EH, Kao YR, Lin CA, Kuo TY, Yang SP, Hsu CF, Chou TY, Ho CC, Wu CW. Hedgehog pathway maintains cell survival under stress conditions, and drives drug resistance in lung adenocarcinoma. Oncotarget 2016; 7:24179-93. [PMID: 27015549 PMCID: PMC5029693 DOI: 10.18632/oncotarget.8253] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 03/02/2016] [Indexed: 01/06/2023] Open
Abstract
Hedgehog (HH) pathway plays an important role in embryonic development, but is largely inactive in adult except for tissue repair. Aberrant activation of HH pathway has been found in a variety of cancer types. In non-small cell lung cancer, however, the role and importance of HH pathway remain controversial. In the current study, we found that HH pathway was maintained in low activity in lung adenocarcinoma (LAC) cells under normal culture condition, but was highly induced in response to stress conditions. Activation of HH pathway promoted cell survival, growth, and invasion partially through HGF and MET signaling. Hedgehog-Interacting Protein (HHIP), a cell-surface negative regulator of HH pathway, was epigenetically silenced in LAC. Overexpression of HHIP blocked the activation of HH and HGF/MET pathways, and made cells significantly more susceptible to stress conditions. In LAC cells with acquired resistance to Epidermal Growth Factor Receptor Tyrosin Kinase Inhibitor (EGFR-TKI), we found that a part of tumor cells were much more sensitive to HH or HGF/MET inhibitors, suggesting an oncogenic addiction shift from EGFR to HH and HGF/MET pathways. In conclusion, this study showed that HH pathway is a survival signaling that drives LAC cell growth under stress conditions, and HHIP is a key regulator to block the induction of HH pathway. Targeting the HH pathway through inhibitors or HHIP thus holds promise to address EGFR-TKI resistance in LAC in clinic.
Collapse
Affiliation(s)
- Erh-Hsuan Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Institute of Microbiology and Immunology, National Yang Ming University, Taipei, Taiwan
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
- Institute of Biochemistry and Molecular Biology, National Yang Ming University, Taipei, Taiwan
| | - Yu-Rung Kao
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chih-An Lin
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University Medical College, Taipei, Taiwan
| | - Ting-Yu Kuo
- Institute of Biochemistry and Molecular Biology, National Yang Ming University, Taipei, Taiwan
| | - Sheng-Ping Yang
- Institute of Microbiology and Immunology, National Yang Ming University, Taipei, Taiwan
| | - Chiung-Fang Hsu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Teh-Ying Chou
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chao-Chi Ho
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University Medical College, Taipei, Taiwan
| | - Cheng-Wen Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Institute of Microbiology and Immunology, National Yang Ming University, Taipei, Taiwan
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
- Institute of Biochemistry and Molecular Biology, National Yang Ming University, Taipei, Taiwan
| |
Collapse
|
30
|
Macdonald TJ. Hedgehog Pathway in Pediatric Cancers: They're Not Just for Brain Tumors Anymore. Am Soc Clin Oncol Educ Book 2016:605-9. [PMID: 24451804 DOI: 10.14694/edbook_am.2012.32.61] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The Hedgehog (HH) pathway regulates fundamental processes in embryonic development, including stem cell maintenance, cell differentiation, tissue polarity, and cell proliferation. In the vertebrate pathway, Sonic hedgehog (SHH) binds to Patched1 (PTCH1), which relieves its inhibition of Smoothened (SMO), allowing the GLI family of transcription factors to translocate to the nucleus and activate HH target genes such as GLI1, GLI2, PTCH1, CYCLIN D1, BCL-2, and MYCN. The HH pathway is also an active participant in tumorigenesis. In 1996, loss-of-function mutation in PTCH1 was discovered to be the cause of nevoid basal cell carcinoma syndrome (NBCCS, or Gorlin syndrome), an autosomal dominant disease associated with increased rates of basal cell carcinoma (BCC), medulloblastoma (MB), and rarely, rhabdomyosarcoma. It is now estimated that 100% of sporadic BCC and up to 20% to 30% of MB also harbor activating HH pathway mutations. Together, these discoveries firmly established the linkage between HH pathway activation and cancer development. Intense research has since been focused on further defining the role of the HH pathway in BCC and MB and potential therapeutic strategies to inhibit HH signaling. Early clinical trials of SMO inhibitors have shown promising results in the treatment of adult BCC and SHH-driven MB. More recently, a number of other pediatric cancers have been reported to show HH activity, making these tumors potential candidates for HH inhibitor therapy. To date however, no HH pathway mutations have been identified in other pediatric cancers. This review will describe the HH pathway signaling in development and cancer with a focus on recent evidence for HH pathway activation in central nervous system (CNS) and non-CNS pediatric cancers.
Collapse
Affiliation(s)
- Tobey J Macdonald
- From the Pediatric Neuro-Oncology Program, Aflac Cancer Center and Blood Disorders Service, Children's Healthcare of Atlanta, and Emory University School of Medicine, Emory Children's Center, Atlanta, GA
| |
Collapse
|
31
|
Tao L, Song C, Huo C, Sun Y, Zhang C, Li X, Yu S, Sun M, Jin B, Zhang Z, Yang K. Anti-CD155 and anti-CD112 monoclonal antibodies conjugated to a fluorescent mesoporous silica nanosensor encapsulating rhodamine 6G and fluorescein for sensitive detection of liver cancer cells. Analyst 2016; 141:4933-40. [DOI: 10.1039/c5an01908g] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Sensitive detection of liver cancer cells using anti-CD155 and anti-CD112 monoclonal antibodies conjugated to ultrabright fluorescent mesoporous silica nanoparticles (FMSNs) encapsulating Rhodamine 6G and fluorescein was developed.
Collapse
Affiliation(s)
- Liang Tao
- Department of Immunology
- The Fourth Military Medical University
- China
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province
- School of Chemistry and Chemical Engineering
| | - Chaojun Song
- Department of Immunology
- The Fourth Military Medical University
- China
| | - Chenyang Huo
- Brigade of Cadet
- The Fourth Military Medical University
- China
| | - Yuanjie Sun
- Department of Immunology
- The Fourth Military Medical University
- China
| | - Chunmei Zhang
- Department of Immunology
- The Fourth Military Medical University
- China
| | - Xiaohua Li
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province
- School of Chemistry and Chemical Engineering
- Shaanxi Normal University
- China
| | - Shaojuan Yu
- Department of Cardiology
- First Hospital of Xi'an
- China
| | - Mingyu Sun
- Brigade of Cadet
- The Fourth Military Medical University
- China
| | - Boquan Jin
- Department of Immunology
- The Fourth Military Medical University
- China
| | - Zhujun Zhang
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province
- School of Chemistry and Chemical Engineering
- Shaanxi Normal University
- China
| | - Kun Yang
- Department of Immunology
- The Fourth Military Medical University
- China
| |
Collapse
|
32
|
Gödeke J, Luxenburger E, Trippel F, Becker K, Häberle B, Müller-Höcker J, von Schweinitz D, Kappler R. Low expression of N-myc downstream-regulated gene 2 (NDRG2) correlates with poor prognosis in hepatoblastoma. Hepatol Int 2015; 10:370-6. [DOI: 10.1007/s12072-015-9686-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 11/06/2015] [Indexed: 12/29/2022]
|
33
|
Garnier A, Ilmer M, Becker K, Häberle B, VON Schweinitz D, Kappler R, Berger M. Truncated neurokinin-1 receptor is an ubiquitous antitumor target in hepatoblastoma, and its expression is independent of tumor biology and stage. Oncol Lett 2015; 11:870-878. [PMID: 26870298 DOI: 10.3892/ol.2015.3951] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 08/25/2015] [Indexed: 12/20/2022] Open
Abstract
The substance P (SP; also known as TAC1)/neurokinin-1 receptor (NK1R; also known as TACR1) complex is a critical part in the development of cancer. Therefore, NK1R antagonists, such as the clinical drug aprepitant, are currently under investigation as future anticancer agents. In a previous study, NK1R (TACR1) was identified as a potent anticancer target in hepatoblastoma (HB). However, little is known regarding the exact distribution of this target among HB subsets and whether it correlates with clinical features and prognosis. In the present study, mRNA was isolated from 47 children with HB, and reverse transcription-quantitative polymerase chain reaction was performed on the samples to analyze the expression of full-length-TACR1 (fl-TACR1) and truncated-TACR1 (tr-TACR1). These data were correlated with data obtained from 9 tumor-free controls, as well as with the presence of metastasis, PRETEXT, vascular invasion, histology, age of diagnosis, multifocality, CTNNB1 mutation, gender and overall survival. Additionally, the present study investigated a recently described 16-gene signature characterizing HB known to correlate with prognosis. Compared with tumor-free liver tissue, tumorous tissue expressed TACR1 significantly higher for the truncated version (P=0.0301), and by trend also for the full-length version. Accordingly, the expression of fl-TACR1 correlated with the expression of the truncated version (P=0.0074). Furthermore, a low expression of fl-TACR1 correlated with characteristics of the 16-gene signature known to predict prognosis (P=0.0222). However, there was no correlation between tr-TACR1 and the tumor characteristics investigated, including outcome, although a clear trend was observed for some tumor characteristics. The current results reinforced the previously described findings that in HB, tr-TACR1 is overexpressed compared with tumor-free liver tissue. Furthermore, to the best of our knowledge, the present study demonstrated for the first time that tr-TACR1 is expressed ubiquitously among the different subsets of HB. Therefore, NK1R may serve as a potent anticancer target in a large variety of patients with HB, independent of tumor biology and clinical stage.
Collapse
Affiliation(s)
- Agnès Garnier
- Department of Pediatric Surgery, Dr von Hauner Children's Hospital, Ludwig Maximilian University of Munich, D-80337 Munich, Germany
| | - Matthias Ilmer
- Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, Hospital of the Ludwig Maximilian University of Munich, D-80336 Munich, Germany
| | - Kristina Becker
- Department of Pediatric Surgery, Dr von Hauner Children's Hospital, Ludwig Maximilian University of Munich, D-80337 Munich, Germany
| | - Beate Häberle
- Department of Pediatric Surgery, Dr von Hauner Children's Hospital, Ludwig Maximilian University of Munich, D-80337 Munich, Germany
| | - Dietrich VON Schweinitz
- Department of Pediatric Surgery, Dr von Hauner Children's Hospital, Ludwig Maximilian University of Munich, D-80337 Munich, Germany
| | - Roland Kappler
- Department of Pediatric Surgery, Dr von Hauner Children's Hospital, Ludwig Maximilian University of Munich, D-80337 Munich, Germany
| | - Michael Berger
- Department of Pediatric Surgery, Dr von Hauner Children's Hospital, Ludwig Maximilian University of Munich, D-80337 Munich, Germany
| |
Collapse
|
34
|
Bhusari S, Pandiri AR, Nagai H, Wang Y, Foley J, Hong HHL, Ton TV, DeVito M, Shockley KR, Peddada SD, Gerrish KE, Malarkey DE, Hooth MJ, Sills RC, Hoenerhoff MJ. Genomic Profiling Reveals Unique Molecular Alterations in Hepatoblastomas and Adjacent Hepatocellular Carcinomas in B6C3F1 Mice. Toxicol Pathol 2015; 43:1114-26. [PMID: 26289556 DOI: 10.1177/0192623315599853] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The cell of origin of hepatoblastoma (HB) in humans and mice is unknown; it is hypothesized to be a transformed hepatocyte, oval cell, or hepatic progenitor cell. In mice, current dogma is that HBs arise from preexisting hepatocellular neoplasms as a result of further neoplastic transformation. However, there is little evidence supporting this direct relationship. To better understand the relationship between hepatocellular carcinoma (HCC) and HB and determine molecular similarities between mouse and human HB, global gene expression analysis and targeted mutation analysis were performed using HB, HCC, and adjacent liver from the same animals in a recent National Toxicology Program bioassay. There were significant differences in Hras and Ctnnb1 mutation spectra, and by microarray, HBs showed dysregulation of embryonic development, stem cell pluripotency, and genomic imprinting compared to HCC. Meta-analysis showed similarities between HB, early mouse embryonic liver, and hepatocyte-derived stem/progenitor cells compared to HCC. Our data show that there are striking differences between HB and HCC and suggest that HB is a significantly different entity that may arise from a hepatic precursor cell. Furthermore, mouse HB is similar to the human disease at the pathway level and therefore is likely a relevant model for evaluating human cancer hazard.
Collapse
Affiliation(s)
- Sachin Bhusari
- Cellular and Molecular Pathology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Arun R Pandiri
- Cellular and Molecular Pathology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA Experimental Pathology Laboratories, Research Triangle Park, North Carolina, USA
| | - Hiroaki Nagai
- Cellular and Molecular Pathology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Yu Wang
- Cellular and Molecular Pathology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Julie Foley
- Cellular and Molecular Pathology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Hue-Hua L Hong
- Cellular and Molecular Pathology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Thai-Vu Ton
- Cellular and Molecular Pathology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Michael DeVito
- Toxicology Branch, Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Keith R Shockley
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Shyamal D Peddada
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Kevin E Gerrish
- Molecular Genomics Core, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - David E Malarkey
- Cellular and Molecular Pathology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Michelle J Hooth
- Program Operations Branch, Division of the National Toxicology Program, Research Triangle Park, North Carolina, USA
| | - Robert C Sills
- Cellular and Molecular Pathology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Mark J Hoenerhoff
- Cellular and Molecular Pathology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| |
Collapse
|
35
|
Shahi MH, Zazpe I, Afzal M, Sinha S, Rebhun RB, Meléndez B, Rey JA, Castresana JS. Epigenetic regulation of human hedgehog interacting protein in glioma cell lines and primary tumor samples. Tumour Biol 2015; 36:2383-91. [PMID: 25416442 PMCID: PMC5012430 DOI: 10.1007/s13277-014-2846-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 11/12/2014] [Indexed: 11/25/2022] Open
Abstract
Glioma constitutes one of the most common groups of brain tumors, and its prognosis is influenced by different genetic and epigenetic modulations. In this study, we demonstrated low or no expression of hedgehog interacting protein (HHIP) in most of the cell lines and primary glioma tumor samples. We further proceeded to promoter methylation study of this gene in the same cell lines and primary tumor samples and found 87 % (7/8) HHIP methylation in glioblastoma cell lines and 75 % (33/44) in primary tumor samples. These methylation pattern correlates with low or unexpressed HHIP in both cell lines and primary tumor samples. Our results suggest the possibility of epigenetic regulation of this gene in glioma, similarly to medulloblastoma, gastric, hepatic, and pancreatic cancers. Also, HHIP might be a diagnostic or prognostic marker in glioma and help to the detection of these tumors in early stages of disease.
Collapse
Affiliation(s)
- Mehdi H. Shahi
- Brain Tumor Biology Unit, University of Navarra School of Sciences, Pamplona, Spain
| | - Idoya Zazpe
- Neurosurgery Service, Hospital of Navarra, Pamplona, Spain
| | - Mohammad Afzal
- Department of Zoology, Aligarh Muslim University, Aligarh, India
| | - Subrata Sinha
- National Brain Research Centre, Manesar, Gurgaon, India
| | - Robert B. Rebhun
- Department of Surgical and Radiological Sciences, University of California Davis School of Veterinary Medicine, Davis, CA, USA
| | - Bárbara Meléndez
- Molecular Pathology Research Unit, Department of Pathology, Virgen de la Salud Hospital, Toledo, Spain
| | - Juan A. Rey
- IdiPaz Research Unit, La Paz University Hospital, Madrid, Spain
| | - Javier S. Castresana
- Brain Tumor Biology Unit, University of Navarra School of Sciences, Pamplona, Spain
| |
Collapse
|
36
|
Fan YH, Ding J, Nguyen S, Liu XJ, Xu G, Zhou HY, Duan NN, Yang SM, Zern MA, Wu J. Aberrant hedgehog signaling is responsible for the highly invasive behavior of a subpopulation of hepatoma cells. Oncogene 2015; 35:116-24. [DOI: 10.1038/onc.2015.67] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 01/14/2015] [Indexed: 02/07/2023]
|
37
|
Eichenmüller M, Trippel F, Kreuder M, Beck A, Schwarzmayr T, Häberle B, Cairo S, Leuschner I, von Schweinitz D, Strom TM, Kappler R. The genomic landscape of hepatoblastoma and their progenies with HCC-like features. J Hepatol 2014; 61:1312-20. [PMID: 25135868 DOI: 10.1016/j.jhep.2014.08.009] [Citation(s) in RCA: 290] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 07/15/2014] [Accepted: 08/07/2014] [Indexed: 02/01/2023]
Abstract
BACKGROUND & AIMS Hepatoblastoma (HB) is the most common childhood liver cancer and occasionally presents with histological and clinical features reminiscent of hepatocellular carcinoma (HCC). Identification of molecular mechanisms that drive the neoplastic continuation towards more aggressive HCC phenotypes may help to guide the new stage of targeted therapies. METHODS We performed comprehensive studies on genetic and chromosomal alterations as well as candidate gene function and their clinical relevance. RESULTS Whole-exome sequencing identified HB as a genetically very simple tumour (2.9 mutations per tumour) with recurrent mutations in ß-catenin (CTNNB1) (12/15 cases) and the transcription factor NFE2L2 (2/15 cases). Their HCC-like progenies share the common CTNNB1 mutation, but additionally exhibit a significantly increased mutation number and chromosomal instability due to deletions of the genome guardians RAD17 and TP53, accompanied by telomerase reverse-transcriptase (TERT) promoter mutations. Targeted genotyping of 33 primary tumours and cell lines revealed CTNNB1, NFE2L2, and TERT mutations in 72.5%, 9.8%, and 5.9% of cases, respectively. All NFE2L2 mutations affected residues of the NFE2L2 protein that are recognized by the KEAP1/CUL3 complex for proteasomal degradation. Consequently, cells transfected with mutant NFE2L2 were insensitive to KEAP1-mediated downregulation of NFE2L2 signalling. Clinically, overexpression of the NFE2L2 target gene NQO1 in tumours was significantly associated with metastasis, vascular invasion, the adverse prognostic C2 gene signature, as well as poor outcome. CONCLUSIONS Our study demonstrates the importance of CTNNB1 mutations and NFE2L2-KEAP1 pathway activation in HB development and defines loss of genomic stability and TERT promoter mutations as prominent characteristics of aggressive HB with HCC features.
Collapse
Affiliation(s)
- Melanie Eichenmüller
- Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Franziska Trippel
- Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Michaela Kreuder
- Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Alexander Beck
- Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Thomas Schwarzmayr
- Institute of Human Genetics, Helmholtz Center Munich, Neuherberg, Germany; Institute of Human Genetics, Technical University Munich, Munich, Germany
| | - Beate Häberle
- Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | | | - Ivo Leuschner
- Institute of Paidopathology, Pediatric Tumor Registry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Dietrich von Schweinitz
- Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Tim M Strom
- Institute of Human Genetics, Helmholtz Center Munich, Neuherberg, Germany; Institute of Human Genetics, Technical University Munich, Munich, Germany
| | - Roland Kappler
- Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University Munich, Munich, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
38
|
Oishi N, Yamashita T, Kaneko S. Molecular biology of liver cancer stem cells. Liver Cancer 2014. [PMID: 24944998 DOI: 10.1159/+000343863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common and lethal cancers worldwide. The concept of cancer stem cells (CSCs) is based primarily on the clinical and experimental observations that indicate the existence of a subpopulation of cells with the capacity to self-renew and differentiate as well as show increased resistance to radiation and chemotherapy. They are considered as the factors responsible for the cases of tumor relapse. Hepatic progenitor cells (HPCs) could form the basis of some hepatocellular carcinomas (HCC) and cholangiocarcinomas. Liver CSCs have been reported in multiple subtypes of HCC and are considered as the master regulators of HCC initiation, tumor metastasis, and progression. HPCs activators such as epithelial cell adhesion molecule (EpCAM), Wnt/β-catenin, transforming growth factor-beta (TGF-β), Notch and Hedgehog signaling systems expedite tumorigenesis or conversely, serve as a powerful cancer-prevention tool. Recent work has also identified Sal-like protein 4 (SALL4) and some epigenetic regulations as important molecules, while several therapeutic drugs that directly control HPCs have been tested both in vivo and in vitro. However, liver CSCs clearly have a complex pathogenesis, with the potential for considerable crosstalk and redundancy in signaling pathways. Hence, the targeting of single molecules or pathways may have limited benefit for treatment. In addition to the direct control of liver CSCs, many other factors are needed for CSC maintenance including angiogenesis, vasculogenesis, invasion and migration, hypoxia, immune evasion, multiple drug resistance, and radioresistance. Here, we provide a brief review of molecular signaling in liver CSCs and present insights into new therapeutic strategies for their targeting.
Collapse
Affiliation(s)
- Naoki Oishi
- Department of Gastroenterology, Kanazawa University Hospital, Ishikawa, Japan
| | - Taro Yamashita
- Department of Gastroenterology, Kanazawa University Hospital, Ishikawa, Japan
| | - Shuichi Kaneko
- Department of Gastroenterology, Kanazawa University Hospital, Ishikawa, Japan
| |
Collapse
|
39
|
Abstract
Hepatocellular carcinoma (HCC) is one of the most common and lethal cancers worldwide. The concept of cancer stem cells (CSCs) is based primarily on the clinical and experimental observations that indicate the existence of a subpopulation of cells with the capacity to self-renew and differentiate as well as show increased resistance to radiation and chemotherapy. They are considered as the factors responsible for the cases of tumor relapse. Hepatic progenitor cells (HPCs) could form the basis of some hepatocellular carcinomas (HCC) and cholangiocarcinomas. Liver CSCs have been reported in multiple subtypes of HCC and are considered as the master regulators of HCC initiation, tumor metastasis, and progression. HPCs activators such as epithelial cell adhesion molecule (EpCAM), Wnt/β-catenin, transforming growth factor-beta (TGF-β), Notch and Hedgehog signaling systems expedite tumorigenesis or conversely, serve as a powerful cancer-prevention tool. Recent work has also identified Sal-like protein 4 (SALL4) and some epigenetic regulations as important molecules, while several therapeutic drugs that directly control HPCs have been tested both in vivo and in vitro. However, liver CSCs clearly have a complex pathogenesis, with the potential for considerable crosstalk and redundancy in signaling pathways. Hence, the targeting of single molecules or pathways may have limited benefit for treatment. In addition to the direct control of liver CSCs, many other factors are needed for CSC maintenance including angiogenesis, vasculogenesis, invasion and migration, hypoxia, immune evasion, multiple drug resistance, and radioresistance. Here, we provide a brief review of molecular signaling in liver CSCs and present insights into new therapeutic strategies for their targeting.
Collapse
Affiliation(s)
| | - Taro Yamashita
- *Taro Yamashita, MD, PhD, Department of Gastroenterology, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8641 (Japan), E-Mail
| | | |
Collapse
|
40
|
Berger M, Neth O, Ilmer M, Garnier A, Salinas-Martín MV, de Agustín Asencio JC, von Schweinitz D, Kappler R, Muñoz M. Hepatoblastoma cells express truncated neurokinin-1 receptor and can be growth inhibited by aprepitant in vitro and in vivo. J Hepatol 2014; 60:985-94. [PMID: 24412605 DOI: 10.1016/j.jhep.2013.12.024] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 11/18/2013] [Accepted: 12/17/2013] [Indexed: 01/12/2023]
Abstract
BACKGROUND & AIMS Multidrug resistance presents a major problem in hepatoblastoma (HB), and new anti-tumor strategies are desperately needed. The substance P (SP)/neurokinin-1 receptor (NK1R) complex has been discovered to be pivotal in the development of a variety of human cancers, and NK1R antagonists, such as the clinical drug aprepitant, are promising future anticancer agents. Yet, the role of the SP/NK1R complex as a potential anticancer target in HB is unknown. METHODS Human HB cell lines HepT1, HepG2, and HuH6, human tumor samples from 17 children with HB as well as mice xenografted with human HB cell line HuH6 were analyzed regarding the SP/NK1R complex as a potential new anti-tumor target in HB. RESULTS Therapeutic targeting with the NK1R antagonists aprepitant, L-733,060, and L-732,138 led to growth inhibition and apoptosis in HepT1, HepG2, and HuH6 cells in a dose-dependent manner. Intriguingly, HB cells predominantly expressed the truncated splice variant of NK1R. Human fibroblasts showed only dismal NK1R expression and were significantly more resistant. Stimulation of HB cells with SP, NK1R's natural ligand, caused increased growth rates and abrogated the anti-proliferative effect of NK1R antagonists. Expression analysis of 17 human HB samples confirmed the clinical relevance of NK1R. Most importantly, oral treatment of a HuH6 xenograft mouse model with 80mg/kg/day aprepitant for 24days resulted in a striking reduction of tumor growth, as evidenced by reduced tumor volume and weight, lowered tumor-specific alpha-fetoprotein (AFP) serum levels, and decreased number of Ki-67 positive cells. Furthermore, aprepitant treatment inhibited in vivo angiogenesis. CONCLUSIONS For the first time, we describe the NK1R in its truncated splice variant as a potent target in human HB and an inhibitory effect in vivo and in vitro by NK1R antagonists. Therefore, NK1R antagonists should be considered promising new candidates for innovative therapeutic strategies against HB.
Collapse
Affiliation(s)
- Michael Berger
- Division of Pediatric Infectious Disease and Immunopathology, Virgen del Rocío Children's Hospital, Institute of Biomedicine, Seville, Spain; Department of Pediatric Surgery, Research Laboratories, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University Munich, Germany
| | - Olaf Neth
- Division of Pediatric Infectious Disease and Immunopathology, Virgen del Rocío Children's Hospital, Institute of Biomedicine, Seville, Spain
| | - Matthias Ilmer
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, University of Texas Health Science Center at Houston, TX, USA
| | - Agnès Garnier
- Department of Pediatric Surgery, Research Laboratories, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University Munich, Germany
| | | | | | - Dietrich von Schweinitz
- Department of Pediatric Surgery, Research Laboratories, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University Munich, Germany
| | - Roland Kappler
- Department of Pediatric Surgery, Research Laboratories, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University Munich, Germany
| | - Miguel Muñoz
- Research Laboratory on Neuropeptides, Virgen del Rocío Children's Hospital, Seville, Spain.
| |
Collapse
|
41
|
López-Terrada D, Alaggio R, de Dávila MT, Czauderna P, Hiyama E, Katzenstein H, Leuschner I, Malogolowkin M, Meyers R, Ranganathan S, Tanaka Y, Tomlinson G, Fabrè M, Zimmermann A, Finegold MJ. Towards an international pediatric liver tumor consensus classification: proceedings of the Los Angeles COG liver tumors symposium. Mod Pathol 2014; 27:472-91. [PMID: 24008558 DOI: 10.1038/modpathol.2013.80] [Citation(s) in RCA: 187] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 02/28/2013] [Accepted: 03/05/2013] [Indexed: 02/07/2023]
Abstract
Liver tumors are rare in children, and their diagnoses may be challenging particularly because of the lack of a current consensus classification system. Systematic central histopathological review of these tumors performed as part of the pediatric collaborative therapeutic protocols has allowed the identification of histologic subtypes with distinct clinical associations. As a result, histopathology has been incorporated within the Children's Oncology Group (COG) protocols, and only in the United States, as a risk-stratification parameter and for patient management. Therefore, the COG Liver Tumor Committee sponsored an International Pathology Symposium in March 2011 to discuss the histopathology and classification of pediatric liver tumors, and hepatoblastoma in particular, and work towards an International Pediatric Liver Tumors Consensus Classification that would be required for international collaborative projects. Twenty-two pathologists and experts in pediatric liver tumors, including those serving as central reviewers for the COG, European Société Internationale d'Oncologie Pédiatrique, Gesellschaft für Pädiatrische Onkologie und Hämatologie, and Japanese Study Group for Pediatric Liver Tumors protocols, as well as pediatric oncologists and surgeons specialized in this field, reviewed more than 50 pediatric liver tumor cases and discussed classic and newly reported entities, as well as criteria for their classification. This symposium represented the first collaborative step to develop a classification that may lead to a common treatment-stratification system incorporating tumor histopathology. A standardized, clinically meaningful classification will also be necessary to allow the integration of new biological parameters and to move towards clinical algorithms based on patient characteristics and tumor genetics, which should improve future patient management and outcome.
Collapse
Affiliation(s)
- Dolores López-Terrada
- Department of Pathology, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Rita Alaggio
- Division of Pathology, Department of Medicine-DIMED, Pathology Unit, Padova, Italy
| | - Maria T de Dávila
- Departamento de Patologia, Hospital de Pediatría Prof. Dr. J.P. Garrahan, Buenos Aires, Argentina
| | - Piotr Czauderna
- Department of Surgery and Urology for Children and Adolescents, Medical University of Gdansk, Gdansk, Poland
| | - Eiso Hiyama
- Department of Surgery, Natural Science Center for Basic Research and Development, Hiroshima University Hospital, Hiroshima, Japan
| | - Howard Katzenstein
- Aflac Cancer Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - Ivo Leuschner
- Institut fur Pathologie, UNI-Klinikum Campus, Kiel, Germany
| | - Marcio Malogolowkin
- Department of Pediatric Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Rebecka Meyers
- Department of Pediatric Surgery, Primary Children's Medical Center, University of Utah, Salt Lake City, UT, USA
| | | | - Yukichi Tanaka
- Division of Pathology, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Gail Tomlinson
- Division of Pediatric Hematology-Oncology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Monique Fabrè
- Department of Pathology, Institut de Cancerologie Gustave Roussy, Villejuif, France
| | | | - Milton J Finegold
- Department of Pathology, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | | |
Collapse
|
42
|
Vengoechea J, Sloan AE, Chen Y, Guan X, Ostrom QT, Kerstetter A, Capella D, Cohen ML, Wolinsky Y, Devine K, Selman W, Barnett GH, Warnick RE, McPherson C, Chiocca EA, Elder JB, Barnholtz-Sloan JS. Methylation markers of malignant potential in meningiomas. J Neurosurg 2013; 119:899-906. [PMID: 23930849 DOI: 10.3171/2013.7.jns13311] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Although most meningiomas are benign, about 20% are atypical (Grade II or III) and have increased mortality and morbidity. Identifying tumors with greater malignant potential can have significant clinical value. This validated genome-wide methylation study comparing Grade I with Grade II and III meningiomas aims to discover genes that are aberrantly methylated in atypical meningiomas. METHODS Patients with newly diagnosed meningioma were identified as part of the Ohio Brain Tumor Study. The Infinium HumanMethylation27 BeadChip (Illumina, Inc.) was used to interrogate 27,578 CpG sites in 14,000 genes per sample for a discovery set of 33 samples (3 atypical). To verify the results, the Infinium HumanMethylation450 BeadChip (Illumina, Inc.) was used to interrogate 450,000 cytosines at CpG loci throughout the genome for a verification set containing 7 replicates (3 atypical), as well as 12 independent samples (6 atypical). A nonparametric Wilcoxon exact test was used to test for difference in methylation between benign and atypical meningiomas in both sets. Heat maps were generated for each set. Methylation results were validated for the 2 probes with the largest difference in methylation intensity by performing Western blot analysis on a set of 20 (10 atypical) samples, including 11 replicates. RESULTS The discovery array identified 95 probes with differential methylation between benign and atypical meningiomas, creating 2 distinguishable groups corresponding to tumor grade when visually examined on a heat map. The validation array evaluated 87 different probes and showed that 9 probes were differentially methylated. On heat map examination the results of this array also suggested the existence of 2 major groups that corresponded to histological grade. IGF2BP1 and PDCD1, 2 proteins that can increase the malignant potential of tumors, were the 2 probes with the largest difference in intensity, and for both of these the atypical meningiomas had a decreased median production of protein, though this was not statistically significant (p = 0.970 for IGF2BP1 and p = 1 for PDCD1). CONCLUSIONS A genome-wide methylation analysis of benign and atypical meningiomas identified 9 genes that were reliably differentially methylated, with the strongest difference in IGF2BP1 and PDCD1. The mechanism why increased methylation of these sites is associated with an aggressive phenotype is not evident. Future research may investigate this mechanism, as well as the utility of IGF2BP1 as a marker for pathogenicity in otherwise benign-appearing meningiomas.
Collapse
Affiliation(s)
- Jaime Vengoechea
- Division of Genetics, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Jeng KS, Sheen IS, Jeng WJ, Yu MC, Hsiau HI, Chang FY, Tsai HH. Activation of the sonic hedgehog signaling pathway occurs in the CD133 positive cells of mouse liver cancer Hepa 1-6 cells. Onco Targets Ther 2013; 6:1047-55. [PMID: 23950652 PMCID: PMC3741084 DOI: 10.2147/ott.s44828] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background The important role of cancer stem cells in carcinogenesis has been emphasized in research. CD133+ cells have been mentioned as liver cancer stem cells in hepatocellular carcinoma (HCC). Some researchers have proposed that the sonic hedgehog (Shh) pathway contributes to hepatocarcinogenesis and that the pathway activation occurs mainly in cancer stem cells. We investigated whether the activation of the Shh pathway occurs in CD133+ cells from liver cancer. Materials and methods We used magnetic sorting to isolate CD133+ cells from mouse cancer Hepa 1–6 cells. To examine the clonogenicity, cell culture and soft agar colony formation assay were performed between CD133+ and CD133− cells. To study the activation of the Shh pathway, we examined the mRNA expressions of Shh, patched homolog 1 (Ptch-1), glioma-associated oncogene homolog 1 (Gli-1), and smoothened homolog (Smoh) by real-time polymerase chain reaction of both CD133+ and CD133− cells. Results The number (mean ± standard deviation) of colonies of CD133+ cells and CD133− cells was 1,031.0 ± 104.7 and 119.7 ± 17.6 respectively. This difference was statistically significant (P < 0.001). Their clonogenicity was 13.7% ± 1.4% and 1.6% ± 0.2% respectively with a statistically significant difference found (P < 0.001). CD133+ cells and CD133− cells were found to have statistically significant differences in Shh mRNA and Smoh mRNA (P = 0.005 and P = 0.043 respectively). Conclusion CD133+ Hepa 1–6 cells have a significantly higher colony proliferation and clonogenicity. The Shh pathway is activated in these cells that harbor stem cell features, with an underexpression of Shh mRNA and an overexpression of Smoh mRNA. Blockade of the Shh signaling pathway may be a potential therapeutic strategy for hepatocarcinogenesis.
Collapse
Affiliation(s)
- Kuo-Shyang Jeng
- Department of Surgery, Far Eastern Memorial Hospital, Taipei, Taiwan, Republic of China
| | | | | | | | | | | | | |
Collapse
|
44
|
El Khatib M, Kalnytska A, Palagani V, Kossatz U, Manns MP, Malek NP, Wilkens L, Plentz RR. Inhibition of hedgehog signaling attenuates carcinogenesis in vitro and increases necrosis of cholangiocellular carcinoma. Hepatology 2013; 57:1035-45. [PMID: 23172661 DOI: 10.1002/hep.26147] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 10/01/2012] [Indexed: 12/22/2022]
Abstract
UNLABELLED The Hedgehog signaling pathway plays a pivotal role during embryonic development, stem cell maintenance, and wound healing. Hedgehog signaling also is deregulated in many cancers. However, the role of this signaling pathway in the carcinogenesis of cholangiocarcinoma (CCC) is still unknown. In this study, we investigated the effects of Hedgehog inhibition by cyclopamine and 5E1 in cultured human CCC cell lines and in vivo using a xenograft mouse model. We also investigated the involvement of Hedgehog in epithelial to mesenchymal transition (EMT), migration, and CCC tumor growth. Sonic hedgehog (Shh) ligand was highly expressed in 89% of human CCC tissues and in CCC cell lines. Cyclopamine and 5E1 treatments effectively inhibited cell proliferation, migration, and invasion by down-regulating the Hedgehog target genes glioblastoma 1 and glioblastoma 2. In vitro and in vivo, we detected an increase in epithelial marker, E-cadherin, after Hedgehog inhibition. In addition, we saw an increase in necrotic areas and a decrease in mitotic figures in cyclopamine and 5E1-treated CCC xenograft tumors. CONCLUSION This study supports the presence of autocrine Hedgehog signaling in human CCC, where CCC cells produce and respond to Shh ligand. Blocking the Hedgehog pathway inhibited EMT and decreased the viability of CCC cells. In addition, cyclopamine and 5E1 inhibited the growth of CCC xenograft tumors.
Collapse
Affiliation(s)
- Mona El Khatib
- Department of Internal Medicine I, Medical University Hospital, Tuebingen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
45
|
López-Terrada D, Zimmermann A. Current issues and controversies in the classification of pediatric hepatocellular tumors. Pediatr Blood Cancer 2012; 59:780-4. [PMID: 22648938 DOI: 10.1002/pbc.24214] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 05/08/2012] [Indexed: 02/03/2023]
Abstract
Systematic histopathologic examination of hepatoblastoma specimens from patients enrolled in therapeutic protocols has allowed the identification of clinically relevant histologic subtypes that are being incorporated into risk stratification systems. Genetic and molecular studies have documented recurrent chromosomal abnormalities and aberrant activation of developmental, and oncogenic signaling pathways in hepatoblastoma. Molecular profiling has also identified molecular subclasses and gene signatures that could be used to stratify hepatoblastoma patients. Future international collaboration is needed to develop consensus pathology classifications, and to progressively incorporate genetic and molecular biomarkers into therapeutic pediatric liver tumors protocols.
Collapse
Affiliation(s)
- Dolores López-Terrada
- Department of Pathology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas 77030, USA.
| | | |
Collapse
|
46
|
Tomlinson GE, Kappler R. Genetics and epigenetics of hepatoblastoma. Pediatr Blood Cancer 2012; 59:785-92. [PMID: 22807084 DOI: 10.1002/pbc.24213] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 05/08/2012] [Indexed: 12/23/2022]
Abstract
A number of unique genetic features are observed in hepatoblastoma that have provided insights into the origins of hepatoblastoma. Hallmark cytogenetic changes in hepatoblastoma include the acquisition of additional copies of whole chromosomes and a recurring unbalanced translocation involving 1q. Genetic syndromes are associated with approximately 15% of hepatoblastoma and the understanding and recognition of these syndromes will be important in determining future surveillance studies needed to prevent additional cancers in survivors as well as in some case guide the care of family members. This article will review the genetic changes, both germ line and acquired, that are recurring events in hepatoblastoma, with emphasis on how these genetic changes could work together with other developmental factors and influence cancer predisposition, tumor growth, as well as aid in prognosis. Tumor-specific signatures based on transcriptional or epigenetic alterations will be reviewed that might be used in the future to better diagnose and subtype the disease as well as predict prognosis and response to therapy.
Collapse
Affiliation(s)
- Gail E Tomlinson
- Department of Pediatrics, Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229, USA.
| | | |
Collapse
|
47
|
SONG KAI, WU JUNHUA, JIANG CHUNPING. Dysregulation of signaling pathways and putative biomarkers in liver cancer stem cells (Review). Oncol Rep 2012; 29:3-12. [DOI: 10.3892/or.2012.2082] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Accepted: 09/25/2012] [Indexed: 02/06/2023] Open
|
48
|
Jeng KS, Sheen IS, Jeng WJ, Yu MC, Tsai HH, Chang FY, Su JC. Blockade of the sonic hedgehog pathway effectively inhibits the growth of hepatoma in mice: An in vivo study. Oncol Lett 2012. [PMID: 23205113 DOI: 10.3892/ol.2012.935] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Hepatocellular carcinoma (HCC), a worldwide malignancy, is prevalent in Asian countries. For individuals with unresectable HCC, the effect of chemotherapy or the present target therapy is limited. There is an urgent need to find innovative new therapies. It is believed that sonic hedgehog (Shh) pathway activation may be essential for hepatocarcinogenesis. In the present study, we conducted an in vivo animal study using an Shh pathway inhibitor to elucidate the effect of treatment upon mice with HCC. Eighty C57BL/6 mice were divided into 4 groups (groups A, B, C and D, with group A serving as a control; n=20 for each). We injected mouse hepatoma Mistheton Lectin-1 cells (5×10(6) cells/20 μl) into the left liver of each mouse in groups B, C and D. In the second week, we analyzed each mouse to assess the tumor growth status. Following the tumor injection, group B did not receive any additional intraperitoneal (i.p.) injection, group C received cyclopamine 10 mg/kg/day i.p. and group D received cyclopamine 30 mg/kg/day i.p. every day for 10 days. After an interval of 4 weeks, harvesting and analysis of the liver was performed for each mouse. Tumor size measurement and real-time PCR of Shh pathway factors (Shh, Ptch-1, Gli-1 and Smoh) for livers of group A and tumors of group B, C and D were undertaken. The decrease in the tumor size of group D was found to be statistically significant (P= 0.047) when compared with groups B or C. The decrease of Shh mRNA of both groups C and D had borderline significance when compared with group B. However, Gli-1 mRNA of group D has statistically significant difference (P=0.044) when compared with group A, B or C. Inhibition of the Shh pathway significantly decreases the size and Gli-1 mRNA expression of the tumor. The Shh pathway may be an effective treatment target for HCC in the future.
Collapse
|
49
|
Guo X, Schmitz JC, Kenney BC, Uchio EM, Kulkarni S, Cha CH. Intermedin is overexpressed in hepatocellular carcinoma and regulates cell proliferation and survival. Cancer Sci 2012; 103:1474-80. [PMID: 22625651 PMCID: PMC7659195 DOI: 10.1111/j.1349-7006.2012.02341.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Revised: 05/03/2012] [Accepted: 05/10/2012] [Indexed: 01/01/2023] Open
Abstract
Angiogenesis is one of the hallmarks of tumor growth and metastasis. Identification of tumor angiogenic factors has been a critical component in understanding cancer biology and treatment. Intermedin (IMD) has been reported to promote angiogenesis in a rat ischemic model and human umbilical vascular endothelial cells. Our study sought to determine the role of IMD in human hepatocellular carcinoma tumor progression. High IMD mRNA expression levels were observed in human hepatocellular carcinoma tumors, even in early stage disease, by real-time RT-PCR. Immunohistochemical analysis of hepatocellular carcinoma clinical samples demonstrated that the tumor regions were significantly more immunoreactive for IMD than adjacent benign liver. Inhibition of IMD expression using RNA interference reduced cell proliferation in SK-Hep-1 and SNU-398 cells. Blockage of IMD signaling using either an antagonist peptide or a neutralizing antibody inhibited growth in a dose-dependent manner with concomitant induction of apoptosis, causing cleavage of caspase-8 and downregulation of Gli1 and Bcl2. Conversely, addition of IMD active peptide increased the phosphorylation level of extracellular signal-regulated kinase. Thus, IMD might play an important role in cell proliferation and survival of hepatocellular carcinoma. Our data suggests that IMD is a potential biomarker and therapeutic target for hepatocellular carcinoma.
Collapse
Affiliation(s)
- Xiaojia Guo
- Department of Surgery, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | | | | | | | | |
Collapse
|
50
|
Venkatramani R, Furman WL, Fuchs J, Warmann SW, Malogolowkin MH. Current and future management strategies for relapsed or progressive hepatoblastoma. Paediatr Drugs 2012; 14:221-32. [PMID: 22702740 DOI: 10.2165/11597740-000000000-00000] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Hepatoblastoma is the most common primary malignant neoplasm of the liver in children. Improvements in chemotherapy and surgical techniques have increased survival rates for those with localized disease. The prognosis for patients with progressive or relapsed disease continues to be dismal. Complete resection by surgery or liver transplantation is necessary for cure. Few conventional chemotherapy agents have demonstrated activity in progressive or relapsed hepatoblastoma. Irinotecan has shown activity in relapsed and progressive hepatoblastoma. The efficacy of high-dose chemotherapy in this setting is unknown. Newer targeted agents that 'selectively' interfere with pathway targets involved in tumor growth and progression such as insulin-like growth factor, phosphatidylinositol 3-kinase (PI3K), Akt, and mammalian target of rapamycin (mTOR) are currently under development. Because of the rarity of hepatoblastoma, only a small minority of these agents will ever be evaluated in children with this disorder. Gene-directed therapy and immunotherapy have shown promising results in the preclinical setting, and should be investigated as future treatment options for advanced hepatoblastoma.
Collapse
|