1
|
Chiu V, Yee C, Main N, Stevanovski I, Watt M, Wilson T, Angus P, Roberts T, Shackel N, Herath C. Oncogenic plasmid DNA and liver injury agent dictates liver cancer development in a mouse model. Clin Sci (Lond) 2024; 138:1227-1248. [PMID: 39254423 PMCID: PMC11427747 DOI: 10.1042/cs20240560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 08/30/2024] [Accepted: 09/10/2024] [Indexed: 09/11/2024]
Abstract
Primary liver cancer is an increasing problem worldwide and is associated with significant mortality. A popular method of modeling liver cancer in mice is plasmid hydrodynamic tail vein injection (HTVI). However, plasmid-HTVI models rarely recapitulate the chronic liver injury which precedes the development of most human liver cancer. We sought to investigate how liver injury using thioacetamide contributes to the pathogenesis and progression of liver cancer in two oncogenic plasmid-HTVI-induced mouse liver cancer models. Fourteen-week-old male mice received double-oncogene plasmid-HTVI (SB/AKT/c-Met and SB/AKT/NRas) and then twice-weekly intraperitoneal injections of thioacetamide for 6 weeks. Liver tissue was examined for histopathological changes, including fibrosis and steatosis. Further characterization of fibrosis and inflammation was performed with immunostaining and real-time quantitative PCR. RNA sequencing with pathway analysis was used to explore novel pathways altered in the cancer models. Hepatocellular and cholangiocellular tumors were observed in mice injected with double-oncogene plasmid-HTVI models (SB/AKT/c-Met and SB/AKT/NRas). Thioacetamide induced mild fibrosis and increased alpha smooth muscle actin-expressing cells. However, the combination of plasmids and thioacetamide did not significantly increase tumor size, but increased multiplicity of small neoplastic lesions. Cancer and/or liver injury up-regulated profibrotic and proinflammatory genes while metabolic pathway genes were mostly down-regulated. We conclude that the liver injury microenvironment can interact with liver cancer and alter its presentation. However, the effects on cancer development vary depending on the genetic drivers with differing active oncogenic pathways. Therefore, the choice of plasmid-HTVI model and injury agent may influence the extent to which injury promotes liver cancer development.
Collapse
Affiliation(s)
- Vincent Chiu
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
- South Western Sydney Clinical School, UNSW Sydney, Liverpool, New South Wales, Australia
| | - Christine Yee
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
- South Western Sydney Clinical School, UNSW Sydney, Liverpool, New South Wales, Australia
| | - Nathan Main
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
- South Western Sydney Clinical School, UNSW Sydney, Liverpool, New South Wales, Australia
| | - Igor Stevanovski
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
- South Western Sydney Clinical School, UNSW Sydney, Liverpool, New South Wales, Australia
| | - Matthew Watt
- School of Biomedical Sciences, University of Melbourne, Victoria, Australia
| | - Trevor Wilson
- Hudson Institute of Medical Research, Monash University, Victoria, Australia
| | - Peter Angus
- Department of Gastroenterology and Hepatology, Austin Health, Heidelberg, Victoria, Australia
| | - Tara Roberts
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Nicholas Shackel
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
- South Western Sydney Clinical School, UNSW Sydney, Liverpool, New South Wales, Australia
| | - Chandana Herath
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
- South Western Sydney Clinical School, UNSW Sydney, Liverpool, New South Wales, Australia
- Department of Medicine, Austin Health, University of Melbourne, Victoria, Australia
| |
Collapse
|
2
|
Shi Q, Xue C, Zeng Y, Yuan X, Chu Q, Jiang S, Wang J, Zhang Y, Zhu D, Li L. Notch signaling pathway in cancer: from mechanistic insights to targeted therapies. Signal Transduct Target Ther 2024; 9:128. [PMID: 38797752 PMCID: PMC11128457 DOI: 10.1038/s41392-024-01828-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/31/2024] [Accepted: 04/15/2024] [Indexed: 05/29/2024] Open
Abstract
Notch signaling, renowned for its role in regulating cell fate, organ development, and tissue homeostasis across metazoans, is highly conserved throughout evolution. The Notch receptor and its ligands are transmembrane proteins containing epidermal growth factor-like repeat sequences, typically necessitating receptor-ligand interaction to initiate classical Notch signaling transduction. Accumulating evidence indicates that the Notch signaling pathway serves as both an oncogenic factor and a tumor suppressor in various cancer types. Dysregulation of this pathway promotes epithelial-mesenchymal transition and angiogenesis in malignancies, closely linked to cancer proliferation, invasion, and metastasis. Furthermore, the Notch signaling pathway contributes to maintaining stem-like properties in cancer cells, thereby enhancing cancer invasiveness. The regulatory role of the Notch signaling pathway in cancer metabolic reprogramming and the tumor microenvironment suggests its pivotal involvement in balancing oncogenic and tumor suppressive effects. Moreover, the Notch signaling pathway is implicated in conferring chemoresistance to tumor cells. Therefore, a comprehensive understanding of these biological processes is crucial for developing innovative therapeutic strategies targeting Notch signaling. This review focuses on the research progress of the Notch signaling pathway in cancers, providing in-depth insights into the potential mechanisms of Notch signaling regulation in the occurrence and progression of cancer. Additionally, the review summarizes pharmaceutical clinical trials targeting Notch signaling for cancer therapy, aiming to offer new insights into therapeutic strategies for human malignancies.
Collapse
Affiliation(s)
- Qingmiao Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Chen Xue
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yifan Zeng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xin Yuan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Qingfei Chu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Shuwen Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jinzhi Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yaqi Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Danhua Zhu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| |
Collapse
|
3
|
Liu ZL, Chen HH, Zheng LL, Sun LP, Shi L. Angiogenic signaling pathways and anti-angiogenic therapy for cancer. Signal Transduct Target Ther 2023; 8:198. [PMID: 37169756 PMCID: PMC10175505 DOI: 10.1038/s41392-023-01460-1] [Citation(s) in RCA: 342] [Impact Index Per Article: 171.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 03/20/2023] [Accepted: 04/20/2023] [Indexed: 05/13/2023] Open
Abstract
Angiogenesis, the formation of new blood vessels, is a complex and dynamic process regulated by various pro- and anti-angiogenic molecules, which plays a crucial role in tumor growth, invasion, and metastasis. With the advances in molecular and cellular biology, various biomolecules such as growth factors, chemokines, and adhesion factors involved in tumor angiogenesis has gradually been elucidated. Targeted therapeutic research based on these molecules has driven anti-angiogenic treatment to become a promising strategy in anti-tumor therapy. The most widely used anti-angiogenic agents include monoclonal antibodies and tyrosine kinase inhibitors (TKIs) targeting vascular endothelial growth factor (VEGF) pathway. However, the clinical benefit of this modality has still been limited due to several defects such as adverse events, acquired drug resistance, tumor recurrence, and lack of validated biomarkers, which impel further research on mechanisms of tumor angiogenesis, the development of multiple drugs and the combination therapy to figure out how to improve the therapeutic efficacy. Here, we broadly summarize various signaling pathways in tumor angiogenesis and discuss the development and current challenges of anti-angiogenic therapy. We also propose several new promising approaches to improve anti-angiogenic efficacy and provide a perspective for the development and research of anti-angiogenic therapy.
Collapse
Affiliation(s)
- Zhen-Ling Liu
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 210009, Nanjing, China
| | - Huan-Huan Chen
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 210009, Nanjing, China
| | - Li-Li Zheng
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 210009, Nanjing, China
| | - Li-Ping Sun
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 210009, Nanjing, China.
| | - Lei Shi
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 210009, Nanjing, China.
| |
Collapse
|
4
|
Testa U, Pelosi E, Castelli G. Cholangiocarcinoma: Molecular Abnormalities and Cells of Origin. Technol Cancer Res Treat 2023; 22:15330338221128689. [PMID: 36872875 PMCID: PMC9989414 DOI: 10.1177/15330338221128689] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/01/2022] [Accepted: 09/07/2022] [Indexed: 03/07/2023] Open
Abstract
Cholangiocarcinomas (CCAs) are a group of heterogeneous epithelial malignancies that can originate at the level of any location of the biliary tree. These tumors are relatively rare but associated with a high rate of mortality. CCAs are morphologically and molecularly heterogeneous and for their location can be distinguished as intracellular and extracellular, subdivided into perihilar and distal. Recent epidemiological, molecular, and cellular studies have supported that the consistent heterogeneity observed for CCAs may result from the convergence of various key elements mainly represented by risk factors, heterogeneity of the associated molecular abnormalities at genetic and epigenetic levels and by different potential cells of origin. These studies have consistently contributed to better defining the pathogenesis of CCAs and to identify in some instances new therapeutic targets. Although the therapeutic progress were still limited, these observations suggest that a better understanding of the molecular mechanisms underlying CCA in the future will help to develop more efficacious treatment strategies.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Oncology, Istituto Supeirore di Sanità, Rome, Italy
| | - Elvira Pelosi
- Department of Oncology, Istituto Supeirore di Sanità, Rome, Italy
| | - Germana Castelli
- Department of Oncology, Istituto Supeirore di Sanità, Rome, Italy
| |
Collapse
|
5
|
Mancarella S, Gigante I, Serino G, Pizzuto E, Dituri F, Valentini MF, Wang J, Chen X, Armentano R, Calvisi DF, Giannelli G. Crenigacestat blocking notch pathway reduces liver fibrosis in the surrounding ecosystem of intrahepatic CCA viaTGF-β inhibition. J Exp Clin Cancer Res 2022; 41:331. [PMID: 36443822 PMCID: PMC9703776 DOI: 10.1186/s13046-022-02536-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 11/09/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Intrahepatic cholangiocarcinoma (iCCA) is a highly malignant tumor characterized by an intensive desmoplastic reaction due to the exaggerated presence of the extracellular (ECM) matrix components. Liver fibroblasts close to the tumor, activated by transforming growth factor (TGF)-β1 and expressing high levels of α-smooth muscle actin (α-SMA), become cancer-associated fibroblasts (CAFs). CAFs are deputed to produce and secrete ECM components and crosstalk with cancer cells favoring tumor progression and resistance to therapy. Overexpression of Notch signaling is implicated in CCA development and growth. The study aimed to determine the effectiveness of the Notch inhibitor, Crenigacestat, on the surrounding microenvironment of iCCA. METHODS We investigated Crenigacestat's effectiveness in a PDX model of iCCA and human primary culture of CAFs isolated from patients with iCCA. RESULTS In silico analysis of transcriptomic profiling from PDX iCCA tissues treated with Crenigacestat highlighted "liver fibrosis" as one of the most modulated pathways. In the iCCA PDX model, Crenigacestat treatment significantly (p < 0.001) reduced peritumoral liver fibrosis. Similar results were obtained in a hydrodynamic model of iCCA. Bioinformatic prediction of the upstream regulators related to liver fibrosis in the iCCA PDX treated with Crenigacestat revealed the involvement of the TGF-β1 pathway as a master regulator gene showing a robust connection between TGF-β1 and Notch pathways. Consistently, drug treatment significantly (p < 0.05) reduced TGF-β1 mRNA and protein levels in tumoral tissue. In PDX tissues, Crenigacestat remarkably inhibited TGF-β signaling and extracellular matrix protein gene expression and reduced α-SMA expression. Furthermore, Crenigacestat synergistically increased Gemcitabine effectiveness in the iCCA PDX model. In 31 iCCA patients, TGF-β1 and α-SMA were upregulated in the tumoral compared with peritumoral tissues. In freshly isolated CAFs from patients with iCCA, Crenigacestat significantly (p < 0.001) inhibited Notch signaling, TGF-β1 secretion, and Smad-2 activation. Consequently, Crenigacestat also inactivated CAFs reducing (p < 0.001) α-SMA expression. Finally, CAFs treated with Crenigacestat produced less (p < 005) ECM components such as fibronectin, collagen 1A1, and collagen 1A2. CONCLUSIONS Notch signaling inhibition reduces the peritumoral desmoplastic reaction in iCCA, blocking the TGF-β1 canonical pathway.
Collapse
Affiliation(s)
- Serena Mancarella
- grid.489101.50000 0001 0162 6994National Institute of Gastroenterology “S. De Bellis” Research Hospital, Via Turi 27, 70013 Castellana Grotte, BA Italy
| | - Isabella Gigante
- grid.489101.50000 0001 0162 6994National Institute of Gastroenterology “S. De Bellis” Research Hospital, Via Turi 27, 70013 Castellana Grotte, BA Italy
| | - Grazia Serino
- grid.489101.50000 0001 0162 6994National Institute of Gastroenterology “S. De Bellis” Research Hospital, Via Turi 27, 70013 Castellana Grotte, BA Italy
| | - Elena Pizzuto
- grid.489101.50000 0001 0162 6994National Institute of Gastroenterology “S. De Bellis” Research Hospital, Via Turi 27, 70013 Castellana Grotte, BA Italy
| | - Francesco Dituri
- grid.489101.50000 0001 0162 6994National Institute of Gastroenterology “S. De Bellis” Research Hospital, Via Turi 27, 70013 Castellana Grotte, BA Italy
| | - Maria F. Valentini
- grid.7644.10000 0001 0120 3326Department of Emergency and Organ Transplant, University of Bari Medical School, Bari, Italy
| | - Jingxiao Wang
- grid.266102.10000 0001 2297 6811Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA 94143 USA
| | - Xin Chen
- grid.266102.10000 0001 2297 6811Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA 94143 USA
| | - Raffaele Armentano
- grid.489101.50000 0001 0162 6994National Institute of Gastroenterology “S. De Bellis” Research Hospital, Via Turi 27, 70013 Castellana Grotte, BA Italy
| | - Diego F. Calvisi
- grid.7727.50000 0001 2190 5763Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany
| | - Gianluigi Giannelli
- grid.489101.50000 0001 0162 6994National Institute of Gastroenterology “S. De Bellis” Research Hospital, Via Turi 27, 70013 Castellana Grotte, BA Italy
| |
Collapse
|
6
|
Cigliano A, Zhang S, Ribback S, Steinmann S, Sini M, Ament CE, Utpatel K, Song X, Wang J, Pilo MG, Berger F, Wang H, Tao J, Li X, Pes GM, Mancarella S, Giannelli G, Dombrowski F, Evert M, Calvisi DF, Chen X, Evert K. The Hippo pathway effector TAZ induces intrahepatic cholangiocarcinoma in mice and is ubiquitously activated in the human disease. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:192. [PMID: 35655220 PMCID: PMC9164528 DOI: 10.1186/s13046-022-02394-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 05/16/2022] [Indexed: 12/31/2022]
Abstract
Background Intrahepatic cholangiocarcinoma (iCCA) is a highly aggressive primary liver tumor with increasing incidence worldwide, dismal prognosis, and few therapeutic options. Mounting evidence underlines the role of the Hippo pathway in this disease; however, the molecular mechanisms whereby the Hippo cascade contributes to cholangiocarcinogenesis remain poorly defined. Methods We established novel iCCA mouse models via hydrodynamic transfection of an activated form of transcriptional coactivator with PDZ-binding motif (TAZ), a Hippo pathway downstream effector, either alone or combined with the myristoylated AKT (myr-AKT) protooncogene, in the mouse liver. Hematoxylin and eosin staining, immunohistochemistry, electron microscopy, and quantitative real-time RT-PCR were applied to characterize the models. In addition, in vitro cell line studies were conducted to address the growth-promoting roles of TAZ and its paralog YAP. Results Overexpression of TAZ in the mouse liver triggered iCCA development with very low incidence and long latency. In contrast, co-expression of TAZ and myr-AKT dramatically increased tumor frequency and accelerated cancer formation in mice, with 100% iCCA incidence and high tumor burden by 10 weeks post hydrodynamic injection. AKT/TAZ tumors faithfully recapitulated many of the histomolecular features of human iCCA. At the molecular level, the development of the cholangiocellular lesions depended on the binding of TAZ to TEAD transcription factors. In addition, inhibition of the Notch pathway did not hamper carcinogenesis but suppressed the cholangiocellular phenotype of AKT/TAZ tumors. Also, knockdown of YAP, the TAZ paralog, delayed cholangiocarcinogenesis in AKT/TAZ mice without affecting the tumor phenotype. Furthermore, human preinvasive and invasive iCCAs and mixed hepatocellular carcinoma/iCCA displayed widespread TAZ activation and downregulation of the mechanisms protecting TAZ from proteolysis. Conclusions Overall, the present data underscore the crucial role of TAZ in cholangiocarcinogenesis Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02394-2.
Collapse
Affiliation(s)
- Antonio Cigliano
- Institute of Pathology, University of Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg, Germany.,Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Shanshan Zhang
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, 513 Parnassus Avenue, San Francisco, CA, USA.,Department of Pathology, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Silvia Ribback
- Institute of Pathology, University of Greifswald, Greifswald, Germany
| | - Sara Steinmann
- Institute of Pathology, University of Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg, Germany
| | - Marcella Sini
- Experimental Pathology Unit, Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Cindy E Ament
- Institute of Pathology, University of Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg, Germany
| | - Kirsten Utpatel
- Institute of Pathology, University of Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg, Germany
| | - Xinhua Song
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, 513 Parnassus Avenue, San Francisco, CA, USA.,School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Jingxiao Wang
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, 513 Parnassus Avenue, San Francisco, CA, USA.,School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Maria G Pilo
- Institute of Pathology, University of Greifswald, Greifswald, Germany
| | - Fabian Berger
- Institute of Pathology, University of Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg, Germany
| | - Haichuan Wang
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, 513 Parnassus Avenue, San Francisco, CA, USA.,Liver Transplantation Division, Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Junyan Tao
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, 513 Parnassus Avenue, San Francisco, CA, USA
| | - Xiaolei Li
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, 513 Parnassus Avenue, San Francisco, CA, USA.,Department of Thyroid and Breast Surgery, The 960th Hospital of the PLA, Jinan, 250031, China
| | - Giovanni M Pes
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Serena Mancarella
- National Institute of Gastroenterology "S. de Bellis", Research Hospital, Castellana Grotte, Italy
| | - Gianluigi Giannelli
- National Institute of Gastroenterology "S. de Bellis", Research Hospital, Castellana Grotte, Italy
| | - Frank Dombrowski
- Institute of Pathology, University of Greifswald, Greifswald, Germany
| | - Matthias Evert
- Institute of Pathology, University of Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg, Germany
| | - Diego F Calvisi
- Institute of Pathology, University of Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg, Germany
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, 513 Parnassus Avenue, San Francisco, CA, USA.,University of Hawaii Cancer Center, Honolulu, Hawaii, USA
| | - Katja Evert
- Institute of Pathology, University of Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg, Germany.
| |
Collapse
|
7
|
Mancarella S, Serino G, Gigante I, Cigliano A, Ribback S, Sanese P, Grossi V, Simone C, Armentano R, Evert M, Calvisi DF, Giannelli G. CD90 is regulated by notch1 and hallmarks a more aggressive intrahepatic cholangiocarcinoma phenotype. J Exp Clin Cancer Res 2022; 41:65. [PMID: 35172861 PMCID: PMC8851853 DOI: 10.1186/s13046-022-02283-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/09/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Intrahepatic Cholangiocarcinoma (iCCA) is characterized by a strong stromal reaction playing a role in tumor progression. Thymus cell antigen 1 (THY1), also called Cluster of Differentiation 90 (CD90), is a key regulator of cell-cell and cell-matrix interaction. In iCCA, CD90 has been reported to be associated with a poor prognosis. In an iCCA PDX model, we recently found that CD90 was downregulated in mice treated with the Notch γ-secretase inhibitor Crenigacestat. The study aims to investigate the role of CD90 in relation to the NOTCH pathway. METHODS THY1/CD90 gene and protein expression was evaluated in human iCCA tissues and xenograft models by qRT-PCR, immunohistochemistry, and immunofluorescence. Notch1 inhibition was achieved by siRNA. THY1/CD90 functions were investigated in xenograft models built with HuCCT1 and KKU-M213 cell lines, engineered to overexpress or knockdown THY1, respectively. RESULTS CD90 co-localized with EPCAM, showing its epithelial origin. In vitro, NOTCH1 silencing triggered HES1 and THY1 down-regulation. RBPJ, a critical transcriptional regulator of NOTCH signaling, exhibited putative binding sites on the THY1 promoter and bound to the latter, implying CD90 as a downstream NOTCH pathway effector. In vivo, Crenigacestat suppressed iCCA growth and reduced CD90 expression in the PDX model. In the xenograft model, Crenigacestat inhibited tumor growth of HuCCT1 cells transfected to overexpress CD90 and KKU-M213 cells constitutively expressing high levels of CD90, while not affecting the growth of HuCCT1 control cells and KKU-M213 depleted of CD90. In an iCCA cohort, patients with higher expression levels of NOTCH1/HES1/THY1 displayed a significantly shorter survival. CONCLUSIONS iCCA patients with higher NOTCH1/HES1/THY1 expression have the worst prognosis, but they are more likely to benefit from Notch signaling inhibition. These findings represent the scientific rationale for testing NOTCH1 inhibitors in clinical trials, taking the first step toward precision medicine for iCCA.
Collapse
Affiliation(s)
- Serena Mancarella
- National Institute of Gastroenterology "S. de Bellis", Research Hospital, Via Turi 27, 70013, Castellana Grotte, Italy
| | - Grazia Serino
- National Institute of Gastroenterology "S. de Bellis", Research Hospital, Via Turi 27, 70013, Castellana Grotte, Italy
| | - Isabella Gigante
- National Institute of Gastroenterology "S. de Bellis", Research Hospital, Via Turi 27, 70013, Castellana Grotte, Italy
| | - Antonio Cigliano
- Institute of Pathology, University of Regensburg, 93053, Regensburg, Germany
| | - Silvia Ribback
- Institute of Pathology, University of Greifswald, 17489, Greifswald, Germany
| | - Paola Sanese
- National Institute of Gastroenterology "S. de Bellis", Research Hospital, Via Turi 27, 70013, Castellana Grotte, Italy
| | - Valentina Grossi
- National Institute of Gastroenterology "S. de Bellis", Research Hospital, Via Turi 27, 70013, Castellana Grotte, Italy
| | - Cristiano Simone
- National Institute of Gastroenterology "S. de Bellis", Research Hospital, Via Turi 27, 70013, Castellana Grotte, Italy
| | - Raffaele Armentano
- National Institute of Gastroenterology "S. de Bellis", Research Hospital, Via Turi 27, 70013, Castellana Grotte, Italy
| | - Matthias Evert
- Institute of Pathology, University of Regensburg, 93053, Regensburg, Germany
| | - Diego F Calvisi
- Institute of Pathology, University of Regensburg, 93053, Regensburg, Germany
| | - Gianluigi Giannelli
- National Institute of Gastroenterology "S. de Bellis", Research Hospital, Via Turi 27, 70013, Castellana Grotte, Italy.
| |
Collapse
|
8
|
Vanaroj P, Chaijaroenkul W, Na-Bangchang K. Notch signaling in the pathogenesis, progression and identification of potential targets for cholangiocarcinoma (Review). Mol Clin Oncol 2022; 16:66. [PMID: 35154706 PMCID: PMC8825743 DOI: 10.3892/mco.2022.2499] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 01/03/2022] [Indexed: 11/05/2022] Open
Abstract
Cholangiocarcinoma (CCA) is an aggressive type of bile duct cancer that is characterized by a high mortality rate due to its late diagnosis and ineffective treatment. The aim of the present systematic review was to analyze the association between Notch signaling and CCA in terms of its pathogenesis, progression and potential treatment targets. Relevant information was gathered from the PubMed, ScienceDirect and Scopus databases using the search terms 'cholangiocarcinoma' AND 'Notch signaling'. Of the 90 articles identified, 28 fulfilled the eligibility criteria and were included in the analysis. It was concluded that overexpression/upregulation of Notch ligands, such as Jagged1 and Notch receptors (Notch1, Notch2 and Notch3), as well as upregulation of the upstream Notch signaling pathway, promoted CCA development and progression. In addition, downregulation of Notch1 signaling through several possible interventions appears to be a promising strategy for inhibition of CCA development and progression. Therefore, the Notch signaling pathway may be considered as a potential target for CCA control.
Collapse
Affiliation(s)
- Peeranate Vanaroj
- Graduate Program in Bioclinical Sciences, Chulabhorn International College of Medicine, Thammasat University, Pathumthani, 12120 Thailand
| | - Wanna Chaijaroenkul
- Graduate Program in Bioclinical Sciences, Chulabhorn International College of Medicine, Thammasat University, Pathumthani, 12120 Thailand
| | - Kesara Na-Bangchang
- Graduate Program in Bioclinical Sciences, Chulabhorn International College of Medicine, Thammasat University, Pathumthani, 12120 Thailand.,Center of Excellence in Pharmacology and Molecular Biology of Malaria and Cholangiocarcinoma, Thammasat University, Pathumthani, 12120 Thailand
| |
Collapse
|
9
|
Song X, Xu H, Wang P, Wang J, Affo S, Wang H, Xu M, Liang B, Che L, Qiu W, Schwabe RF, Chang TT, Vogl M, Pes GM, Ribback S, Evert M, Chen X, Calvisi DF. Focal adhesion kinase (FAK) promotes cholangiocarcinoma development and progression via YAP activation. J Hepatol 2021; 75:888-899. [PMID: 34052254 PMCID: PMC8453055 DOI: 10.1016/j.jhep.2021.05.018] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 04/28/2021] [Accepted: 05/14/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase that is upregulated in many tumor types and is a promising target for cancer therapy. Herein, we elucidated the functional role of FAK in intrahepatic cholangiocarcinoma (iCCA) development and progression. METHODS Expression levels and activation status of FAK were determined in human iCCA samples. The functional contribution of FAK to Akt/YAP murine iCCA initiation and progression was investigated using conditional Fak knockout mice and constitutive Cre or inducible Cre mice, respectively. The oncogenic potential of FAK was further examined via overexpression of FAK in mice. In vitro cell line studies and in vivo drug treatment were applied to address the therapeutic potential of targeting FAK for iCCA treatment. RESULTS FAK was ubiquitously upregulated and activated in iCCA lesions. Ablation of FAK strongly delayed Akt/YAP-driven mouse iCCA initiation. FAK overexpression synergized with activated AKT to promote iCCA development and accelerated Akt/Jag1-driven cholangiocarcinogenesis. Mechanistically, FAK was required for YAP(Y357) phosphorylation, supporting the role of FAK as a central YAP regulator in iCCA. Significantly, ablation of FAK after Akt/YAP-dependent iCCA formation strongly suppressed tumor progression in mice. Furthermore, a remarkable iCCA growth reduction was achieved when a FAK inhibitor and palbociclib, a CDK4/6 inhibitor, were administered simultaneously in human iCCA cell lines and Akt/YAP mice. CONCLUSIONS FAK activation contributes to the initiation and progression of iCCA by inducing the YAP proto-oncogene. Targeting FAK, either alone or in combination with anti-CDK4/6 inhibitors, may be an effective strategy for iCCA treatment. LAY SUMMARY We found that the protein FAK (focal adhesion kinase) is upregulated and activated in human and mouse intrahepatic cholangiocarcinoma samples. FAK promotes intrahepatic cholangiocarcinoma development, whereas deletion of FAK strongly suppresses its initiation and progression. Combined FAK and CDK4/6 inhibitor treatment had a strong anti-cancer effect in in vitro and in vivo models. This combination therapy might represent a valuable and novel treatment against human intrahepatic cholangiocarcinoma.
Collapse
Affiliation(s)
- Xinhua Song
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA, USA.
| | - Hongwei Xu
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA, USA,Liver Transplantation Division, Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, China; Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Pan Wang
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA, USA,Collaborative Innovation Center for Agricultural Product Processing and Nutrition & Health, Beijing Vegetable Research Center, Beijing Academy of Agriculture and Forestry Science, Beijing, China
| | - Jingxiao Wang
- Beijing University of Chinese Medicine, Beijing, China
| | - Silvia Affo
- Department of Medicine, Columbia University, New York, NY, USA
| | - Haichuan Wang
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA, USA,Liver Transplantation Division, Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, China; Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Meng Xu
- Department of General Surgery, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an Jiaotong University, Xi’an, PR China
| | - Binyong Liang
- Hepatic Surgery Center, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Che
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA, USA
| | - Wei Qiu
- Department of Surgery and Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL
| | | | - Tammy T Chang
- Department of Surgery and Liver Center, University of California, San Francisco, CA, USA
| | - Marion Vogl
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Giovanni M. Pes
- Department of Medical, Surgical, and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Silvia Ribback
- Institute of Pathology, University of Greifswald, Greifswald, Germany
| | - 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, CA, USA.
| | - Diego F. Calvisi
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| |
Collapse
|
10
|
Kamalakar A, McKinney JM, Salinas Duron D, Amanso AM, Ballestas SA, Drissi H, Willett NJ, Bhattaram P, García AJ, Wood LB, Goudy SL. JAGGED1 stimulates cranial neural crest cell osteoblast commitment pathways and bone regeneration independent of canonical NOTCH signaling. Bone 2021; 143:115657. [PMID: 32980561 PMCID: PMC9035226 DOI: 10.1016/j.bone.2020.115657] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/14/2020] [Accepted: 09/16/2020] [Indexed: 12/21/2022]
Abstract
Craniofacial bone loss is a complex clinical problem with limited regenerative solutions. Currently, BMP2 is used as a bone-regenerative therapy in adults, but in pediatric cases of bone loss, it is not FDA-approved due to concerns of life-threatening inflammation and cancer. Development of a bone-regenerative therapy for children will transform our ability to reduce the morbidity associated with current autologous bone grafting techniques. We discovered that JAGGED1 (JAG1) induces cranial neural crest (CNC) cell osteoblast commitment during craniofacial intramembranous ossification, suggesting that exogenous JAG1 delivery is a potential craniofacial bone-regenerative approach. In this study, we found that JAG1 delivery using synthetic hydrogels containing O9-1 cells, a CNC cell line, into critical-sized calvarial defects in C57BL/6 mice provided robust bone-regeneration. Since JAG1 signals through canonical (Hes1/Hey1) and non-canonical (JAK2) NOTCH pathways in CNC cells, we used RNAseq to analyze transcriptional pathways activated in CNC cells treated with JAG1 ± DAPT, a NOTCH-canonical pathway inhibitor. JAG1 upregulated expression of multiple NOTCH canonical pathway genes (Hes1), which were downregulated in the presence of DAPT. JAG1 also induced bone chemokines (Cxcl1), regulators of cytoskeletal organization and cell migration (Rhou), signaling targets (STAT5), promoters of early osteoblast cell proliferation (Prl2c2, Smurf1 and Esrra), and, inhibitors of osteoclasts (Id1). In the presence of DAPT, expression levels of Hes1 and Cxcl1 were decreased, whereas, Prl2c2, Smurf1, Esrra, Rhou and Id1 remain elevated, suggesting that JAG1 induces osteoblast proliferation through these non-canonical genes. Pathway analysis of JAG1 + DAPT-treated CNC cells revealed significant upregulation of multiple non-canonical pathways, including the cell cycle, tubulin pathway, regulators of Runx2 initiation and phosphorylation of STAT5 pathway. In total, our data show that JAG1 upregulates multiple pathways involved in osteogenesis, independent of the NOTCH canonical pathway. Moreover, our findings suggest that JAG1 delivery using a synthetic hydrogel, is a bone-regenerative approach with powerful translational potential.
Collapse
Affiliation(s)
| | - Jay M McKinney
- Wallace H. Coulter Department of Biomedical Engineering, USA; George W. Woodruff School of Mechanical Engineering, Georgia Tech College of Engineering, Atlanta, GA, USA; The Atlanta Veterans Affairs Medical Center Atlanta, GA, USA.
| | | | | | | | - Hicham Drissi
- Department of Cell Biology, USA; Department of Orthopaedics, Emory University, Atlanta, GA, USA; The Atlanta Veterans Affairs Medical Center Atlanta, GA, USA.
| | - Nick J Willett
- Department of Orthopaedics, Emory University, Atlanta, GA, USA; The Atlanta Veterans Affairs Medical Center Atlanta, GA, USA.
| | - Pallavi Bhattaram
- Department of Cell Biology, USA; Department of Orthopaedics, Emory University, Atlanta, GA, USA.
| | - Andrés J García
- Parker H. Petit Institute for Bioengineering and Biosciences, USA; George W. Woodruff School of Mechanical Engineering, Georgia Tech College of Engineering, Atlanta, GA, USA.
| | - Levi B Wood
- George W. Woodruff School of Mechanical Engineering, Georgia Tech College of Engineering, Atlanta, GA, USA.
| | - Steven L Goudy
- Department of Otolaryngology, USA; Department of Pediatric Otolaryngology, Children's Healthcare of Atlanta, Atlanta, GA, USA.
| |
Collapse
|
11
|
Moeini A, Haber PK, Sia D. Cell of origin in biliary tract cancers and clinical implications. JHEP Rep 2021; 3:100226. [PMID: 33665585 PMCID: PMC7902553 DOI: 10.1016/j.jhepr.2021.100226] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 12/12/2022] Open
Abstract
Biliary tract cancers (BTCs) are aggressive epithelial malignancies that can arise at any point of the biliary tree. Albeit rare, their incidence and mortality rates have been rising steadily over the past 40 years, highlighting the need to improve current diagnostic and therapeutic strategies. BTCs show high inter- and intra-tumour heterogeneity both at the morphological and molecular level. Such complex heterogeneity poses a substantial obstacle to effective interventions. It is widely accepted that the observed heterogeneity may be the result of a complex interplay of different elements, including risk factors, distinct molecular alterations and multiple potential cells of origin. The use of genetic lineage tracing systems in experimental models has identified cholangiocytes, hepatocytes and/or progenitor-like cells as the cells of origin of BTCs. Genomic evidence in support of the distinct cell of origin hypotheses is growing. In this review, we focus on recent advances in the histopathological subtyping of BTCs, discuss current genomic evidence and outline lineage tracing studies that have contributed to the current knowledge surrounding the cell of origin of these tumours.
Collapse
Key Words
- ARID1A, AT-rich interactive domain-containing protein 1A
- BAP1, BRCA1-associated protein 1
- BRAF, v-Raf murine sarcoma viral oncogene homolog B
- BTC, biliary tract cancer
- Biliary tract cancers
- CCA, cholangiocarcinoma
- CDKN2A/B, cyclin-dependent kinase inhibitor 2A/B
- CK, cytokeratin
- CLC, cholangiolocarcinoma
- Cell of origin
- Cholangiocarcinoma
- CoH, Canal of Hering
- DCR, disease control rate
- ER, estrogen receptor
- ERBB2/3, Erb-B2 Receptor Tyrosine Kinase 2/3
- FGFR, fibroblast growth factor receptor
- FGFR2, Fibroblast Growth Factor Receptor 2
- GBC, gallbladder cancer
- GEMM, genetically engineered mouse models
- Genomics
- HCC, hepatocellular carcinoma
- HPCs, hepatic progenitor cells
- IDH, isocitrate dehydrogenase
- KRAS, Kirsten Rat Sarcoma Viral Oncogene Homolog
- Lineage tracing
- MET, Hepatocyte Growth Factor Receptor
- MST1, Macrophage Stimulating 1
- NA, not applicable
- NAFLD, non-alcoholic fatty liver disease
- NASH, non-alcoholic steatohepatitis
- NGS, next-generation sequencing
- NR, not reported
- NTRK, Neurotrophic Receptor Tyrosine Kinase 1
- ORR, objective response rate
- OS, overall survival
- PBG, peribiliary gland
- PFS, progression- free survival
- PIK3CA, Phosphatidylinositol-4,5-Bisphosphate 3-Kinase Catalytic Subunit Alpha
- PLC, primary liver cancer
- PRKACA/B, Protein Kinase CAMP-Activated Catalytic Subunit Alpha/Beta
- PROM1, Prominin 1
- PSC, primary sclerosing cholangitis
- Personalized therapy
- RNF43, Ring Finger Protein 43
- SMAD4, SMAD Family Member 4
- TBG, thyroid binding globulin
- TP53, Tumor Protein P53
- WHO, World Health Organization
- dCCA, distal cholangiocarcinoma
- eCCA, extrahepatic cholangiocarcinoma
- iCCA, intrahepatic cholangiocarcinoma
- mo, months
- pCCA, perihilar cholangiocarcinoma
Collapse
Affiliation(s)
- Agrin Moeini
- Cancer Inflammation and Immunity Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, Manchester, UK
| | - Philipp K Haber
- Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Daniela Sia
- Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| |
Collapse
|
12
|
Brandi G, Tavolari S. In Vitro and In Vivo Model Systems of Cholangiocarcinoma. DIAGNOSIS AND MANAGEMENT OF CHOLANGIOCARCINOMA 2021:471-494. [DOI: 10.1007/978-3-030-70936-5_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
13
|
Schindler EA, Gilbert MA, Piccoli DA, Spinner NB, Krantz ID, Loomes KM. Alagille syndrome and risk for hepatocellular carcinoma: Need for increased surveillance in adults with mild liver phenotypes. Am J Med Genet A 2020; 185:719-731. [PMID: 33369123 PMCID: PMC7898517 DOI: 10.1002/ajmg.a.62028] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 11/18/2020] [Accepted: 11/27/2020] [Indexed: 12/26/2022]
Abstract
Alagille syndrome (ALGS) is a multisystem autosomal dominant developmental disorder caused predominantly by pathogenic variants in JAGGED1 (JAG1), and also by pathogenic variants in NOTCH2 in a much smaller number of individuals. Clinical presentation is highly variable and includes liver, heart, eye, skeleton, and facial abnormalities, with a subset of individuals also presenting with kidney, vascular, and central nervous system phenotypes. Hepatocellular carcinoma (HCC) is a rare complication of ALGS, though little is known about its incidence or etiology among affected individuals. Previous reports have identified HCC occurrence in both pediatric and adult cases of ALGS. We present a case report of HCC in a 58‐year‐old woman with a pathogenic JAG1 variant and no overt hepatic features of ALGS. Through a comprehensive literature review, we compile all reported pediatric and adult cases, and further highlight one previously reported case of HCC onset in an adult ALGS patient without any hepatic disease features, similar to our own described patient. Our case report and literature review suggest that ALGS‐causing variants could confer risk for developing HCC regardless of phenotypic severity and highlight a need for a cancer screening protocol that would enable early detection and treatment in this at‐risk population.
Collapse
Affiliation(s)
- Emma A Schindler
- Division of Human Genetics, Roberts Individualized Medical Genetics Center, Children's Hospital of Philadelphia and The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Melissa A Gilbert
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia and The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David A Piccoli
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Children's Hospital of Philadelphia and Department of Pediatrics, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nancy B Spinner
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia and The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ian D Krantz
- Division of Human Genetics, Roberts Individualized Medical Genetics Center, Children's Hospital of Philadelphia and The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kathleen M Loomes
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Children's Hospital of Philadelphia and Department of Pediatrics, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
14
|
Rauff B, Malik A, Bhatti YA, Chudhary SA, Qadri I, Rafiq S. Notch signalling pathway in development of cholangiocarcinoma. World J Gastrointest Oncol 2020; 12:957-974. [PMID: 33005291 PMCID: PMC7509998 DOI: 10.4251/wjgo.v12.i9.957] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/03/2020] [Accepted: 08/25/2020] [Indexed: 02/06/2023] Open
Abstract
Cholangiocarcinoma (CCA) comprises of extra-hepatic cholangiocarcinoma and intrahepatic cholangiocarcinoma cancers as a result of inflammation of epithelium cell lining of the bile duct. The incidence rate is increasing dramatically worldwide with highest rates in Eastern and South Asian regions. Major risk factors involve chronic damage and inflammation of bile duct epithelium from primary sclerosing cholangitis, chronic hepatitis virus infection, gallstones and liver fluke infection. Various genetic variants have also been identified and as CCA develops on the background of biliary inflammation, diverse range of molecular mechanisms are involved in its progression. Among these, the Notch signalling pathway acts as a major driver of cholangiocarcinogenesis and its components (receptors, ligands and downstream signalling molecules) represent a promising therapeutic targets. Gamma-Secretase Inhibitors have been recognized in inhibiting the Notch pathway efficiently. A comprehensive knowledge of the molecular pathways activated by the Notch signalling cascade as well as its functional crosstalk with other signalling pathways provide better approach in developing innovative therapies against CCA.
Collapse
Affiliation(s)
- Bisma Rauff
- Institute of Molecular Biology and Biotechnology, University of Lahore, Lahore 54000, Pakistan
| | - Arif Malik
- Institute of Molecular Biology and Biotechnology, University of Lahore, Lahore 54000, Pakistan
| | - Yasir Ali Bhatti
- Institute of Molecular Biology and Biotechnology, University of Lahore, Lahore 54000, Pakistan
| | - Shafiq Ahmad Chudhary
- Institute of Biomedical and Allied Health Sciences, University of Health Sciences, Lahore 54000, Pakistan
| | - Ishtiaq Qadri
- Department of Biology, Faculty of Science, King Abdulaziz University Jeddah Kingdom of Saudi Arabia
| | - Shafquat Rafiq
- Department of Gastrointestinal medicine, Croydon University Hospital, Croydon CR7 7YE, United Kingdom
| |
Collapse
|
15
|
Zhu Y, Kwong LN. Insights Into the Origin of Intrahepatic Cholangiocarcinoma From Mouse Models. Hepatology 2020; 72:305-314. [PMID: 32096245 DOI: 10.1002/hep.31200] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/17/2020] [Accepted: 02/11/2020] [Indexed: 12/17/2022]
Affiliation(s)
- Yan Zhu
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Lawrence N Kwong
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
16
|
Zhang S, Zhang J, Evert K, Li X, Liu P, Kiss A, Schaff Z, Ament C, Zhang Y, Serra M, Evert M, Chen N, Xu F, Chen X, Tao J, Calvisi DF, Cigliano A. The Hippo Effector Transcriptional Coactivator with PDZ-Binding Motif Cooperates with Oncogenic β-Catenin to Induce Hepatoblastoma Development in Mice and Humans. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:1397-1413. [PMID: 32283103 DOI: 10.1016/j.ajpath.2020.03.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 03/08/2020] [Accepted: 03/12/2020] [Indexed: 02/05/2023]
Abstract
Hepatoblastoma (HB) is the most common pediatric liver tumor. Though Wnt/β-catenin and Hippo cascades are implicated in HB development, studies on crosstalk between β-catenin and Hippo downstream effector transcriptional coactivator with PDZ-binding motif (TAZ) in HB are lacking. Expression levels of TAZ and β-catenin in human HB specimens were assessed by immunohistochemistry. Functional interplay between TAZ and β-catenin was determined by overexpression of an activated form of TAZ (TAZS89A), either alone or combined with an oncogenic form of β-catenin (ΔN90-β-catenin), in mouse liver via hydrodynamic transfection. Activation of TAZ often co-occurred with that of β-catenin in clinical specimens. Although the overexpression of TAZS89A alone did not induce hepatocarcinogenesis, concomitant overexpression of TAZS89A and ΔN90-β-catenin triggered the development of HB lesions exhibiting both epithelial and mesenchymal features. Mechanistically, TAZ/β-catenin-driven HB development required TAZ interaction with transcriptional enhanced associate domain factors. Blockade of the Notch cascade did not inhibit TAZ/β-catenin-dependent HB formation in mice but suppressed the mesenchymal phenotype. Neither Yes-associated protein nor heat shock factor 1 depletion affected HB development in TAZ/β-catenin mice. In human HB cell lines, silencing of TAZ resulted in decreased cell growth, which was further reduced when TAZ knockdown was associated with suppression of either β-catenin or Yes-associated protein. Overall, our study identified TAZ as a crucial oncogene in HB development and progression.
Collapse
Affiliation(s)
- Shu Zhang
- Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, PR China; Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, San Francisco, California
| | - Jie Zhang
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, San Francisco, California; Department of Thoracic Oncology II, Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital and Institute, Beijing, PR China
| | - Katja Evert
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Xiaolei Li
- Department of Thyroid and Breast Surgery, The 960th Hospital of the PLA, Jinan, PR China
| | - Pin Liu
- Department of Pediatrics, Zhongnan Hospital of Wuhan University, Wuhan, PR China
| | - Andras Kiss
- Second Department of Pathology, Semmelweis University, Budapest, Hungary
| | - Zsuzsa Schaff
- Second Department of Pathology, Semmelweis University, Budapest, Hungary
| | - Cindy Ament
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Yi Zhang
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, San Francisco, California; Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Monica Serra
- Department of Medical, Surgical, and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Matthias Evert
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Nianyong Chen
- Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, PR China
| | - Feng Xu
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, PR China
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, San Francisco, California
| | - Junyan Tao
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.
| | - Diego F Calvisi
- Department of Medical, Surgical, and Experimental Sciences, University of Sassari, Sassari, Italy.
| | - Antonio Cigliano
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| |
Collapse
|
17
|
Crenigacestat, a selective NOTCH1 inhibitor, reduces intrahepatic cholangiocarcinoma progression by blocking VEGFA/DLL4/MMP13 axis. Cell Death Differ 2020; 27:2330-2343. [PMID: 32042099 PMCID: PMC7370218 DOI: 10.1038/s41418-020-0505-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 01/16/2020] [Accepted: 01/21/2020] [Indexed: 02/06/2023] Open
Abstract
Intrahepatic cholangiocarcinoma (iCCA) is a deadly disease with rising incidence and few treatment options. An altered expression and/or activation of NOTCH1–3 receptors has been shown to play a role in iCCA development and progression. In this study, we established a new CCA patient-derived xenograft model, which was validated by immunohistochemistry and transcriptomic analysis. The effects of Notch pathway suppression by the Crenigacestat (LY3039478)-specific inhibitor were evaluated in human iCCA cell lines and the PDX model. In vitro, LY3039478 significantly reduced Notch pathway components, including NICD1 and HES1, but not the other Notch receptors, in a panel of five different iCCA cell lines. In the PDX model, LY3039478 significantly inhibited the Notch pathway and tumor growth to the same extent as gemcitabine. Furthermore, gene expression analysis of iCCA mouse tissues treated with LY3039478 revealed a downregulation of VEGFA, HES1, and MMP13 genes. In the same tissues, DLL4 and CD31 co-localized, and their expression was significantly inhibited in the treated mice, as it happened in the case of MMP13. In an in vitro angiogenesis model, LY3039478 inhibited vessel formation, which was restored by the addition of MMP13. Finally, RNA-sequencing expression data of iCCA patients and matched surrounding normal liver tissues downloaded from the GEO database demonstrated that NOTCH1, HES1, MMP13, DLL4, and VEGFA genes were significantly upregulated in tumors compared with adjacent nontumorous tissues. These data were confirmed by our group, using an independent cohort of iCCA specimens. Conclusion: We have developed and validated a new iCCA PDX model to test in vivo the activity of LY3039478, demonstrating its inhibitory role in Notch-dependent angiogenesis. Thus, the present data provide new knowledge on Notch signaling in iCCA, and support the inhibition of the Notch cascade as a promising strategy for the treatment of this disease.
Collapse
|
18
|
Qiao Y, Wang J, Karagoz E, Liang B, Song X, Shang R, Evert K, Xu M, Che L, Evert M, Calvisi DF, Tao J, Wang B, Monga SP, Chen X. Axis inhibition protein 1 (Axin1) Deletion-Induced Hepatocarcinogenesis Requires Intact β-Catenin but Not Notch Cascade in Mice. Hepatology 2019; 70:2003-2017. [PMID: 30737831 PMCID: PMC7206928 DOI: 10.1002/hep.30556] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 02/02/2019] [Indexed: 12/12/2022]
Abstract
Inactivating mutations of axis inhibition protein 1 (AXIN1), a negative regulator of the Wnt/β-Catenin cascade, are among the common genetic events in human hepatocellular carcinoma (HCC), affecting approximately 10% of cases. In the present manuscript, we sought to define the genetic crosstalk between Axin1 mutants and Wnt/β-catenin as well as Notch signaling cascades along hepatocarcinogenesis. We discovered that c-MET activation and AXIN1 mutations occur concomitantly in ~3%-5% of human HCC samples. Subsequently, we generated a murine HCC model by means of CRISPR/Cas9-based gene deletion of Axin1 (sgAxin1) in combination with transposon-based expression of c-Met in the mouse liver (c-Met/sgAxin1). Global gene expression analysis of mouse normal liver, HCCs induced by c-Met/sgAxin1, and HCCs induced by c-Met/∆N90-β-Catenin revealed activation of the Wnt/β-Catenin and Notch signaling in c-Met/sgAxin1 HCCs. However, only a few of the canonical Wnt/β-Catenin target genes were induced in c-Met/sgAxin1 HCC when compared with corresponding lesions from c-Met/∆N90-β-Catenin mice. To study whether endogenous β-Catenin is required for c-Met/sgAxin1-driven HCC development, we expressed c-Met/sgAxin1 in liver-specific Ctnnb1 null mice, which completely prevented HCC development. Consistently, in AXIN1 mutant or null human HCC cell lines, silencing of β-Catenin strongly inhibited cell proliferation. In striking contrast, blocking the Notch cascade through expression of either the dominant negative form of the recombinant signal-binding protein for immunoglobulin kappa J region (RBP-J) or the ablation of Notch2 did not significantly affect c-Met/sgAxin1-driven hepatocarcinogenesis. Conclusion: We demonstrated here that loss of Axin1 cooperates with c-Met to induce HCC in mice, in a β-Catenin signaling-dependent but Notch cascade-independent way.
Collapse
Affiliation(s)
- Yu Qiao
- Department of Oncology, Beijing Hospital, National Center of Gerontology, Beijing, China;,Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA
| | - Jingxiao Wang
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA;,School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Eylul Karagoz
- Department of Medicine and Liver Center, University of California, San Francisco, CA;,School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Binyong Liang
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA;,Hepatic Surgery Center, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinhua Song
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA;,Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Runze Shang
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA;,Department of Hepatobiliary Surgery, Xijing Hospital, Air Force Military Medical University, Xi’an, China
| | - Katja Evert
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Meng Xu
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA;,Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Li Che
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA
| | - Matthias Evert
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Diego F. Calvisi
- Institute of Pathology, University Medicine Greifswald, Greifswald, Germany
| | - Junyan Tao
- Department of Pathology, University of Pittsburgh School of Medicine, and Pittsburgh Liver Research Center, Pittsburgh, PA
| | - Bruce Wang
- Department of Medicine and Liver Center, University of California, San Francisco, CA
| | - Satdarshan P. Monga
- Department of Pathology, University of Pittsburgh School of Medicine, and Pittsburgh Liver Research Center, Pittsburgh, PA
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA
| |
Collapse
|
19
|
Erice O, Vallejo A, Ponz-Sarvise M, Saborowski M, Vogel A, Calvisi DF, Saborowski A, Vicent S. Genetic Mouse Models as In Vivo Tools for Cholangiocarcinoma Research. Cancers (Basel) 2019; 11:cancers11121868. [PMID: 31769429 PMCID: PMC6966555 DOI: 10.3390/cancers11121868] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 11/22/2019] [Indexed: 02/07/2023] Open
Abstract
Cholangiocarcinoma (CCA) is a genetically and histologically complex disease with a highly dismal prognosis. A deeper understanding of the underlying cellular and molecular mechanisms of human CCA will increase our current knowledge of the disease and expedite the eventual development of novel therapeutic strategies for this fatal cancer. This endeavor is effectively supported by genetic mouse models, which serve as sophisticated tools to systematically investigate CCA pathobiology and treatment response. These in vivo models feature many of the genetic alterations found in humans, recapitulate multiple hallmarks of cholangiocarcinogenesis (encompassing cell transformation, preneoplastic lesions, established tumors and metastatic disease) and provide an ideal experimental setting to study the interplay between tumor cells and the surrounding stroma. This review is intended to serve as a compendium of CCA mouse models, including traditional transgenic models but also genetically flexible approaches based on either the direct introduction of DNA into liver cells or transplantation of pre-malignant cells, and is meant as a resource for CCA researchers to aid in the selection of the most appropriate in vivo model system.
Collapse
Affiliation(s)
- Oihane Erice
- Center for Applied Medical Research, Program in Solid Tumors, University of Navarra, 31008 Pamplona, Spain; (O.E.); (A.V.)
| | - Adrian Vallejo
- Center for Applied Medical Research, Program in Solid Tumors, University of Navarra, 31008 Pamplona, Spain; (O.E.); (A.V.)
| | - Mariano Ponz-Sarvise
- Department of Medical Oncology, Clinica Universidad de Navarra, 31008 Pamplona, Spain;
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - Michael Saborowski
- Department of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School, 30625 Hannover, Germany; (M.S.); (A.V.)
| | - Arndt Vogel
- Department of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School, 30625 Hannover, Germany; (M.S.); (A.V.)
| | - Diego F. Calvisi
- Institute for Pathology, Regensburg University, 93053 Regensburg, Germany;
| | - Anna Saborowski
- Department of Medical Oncology, Clinica Universidad de Navarra, 31008 Pamplona, Spain;
- Correspondence: (A.S.); (S.V.); Tel.: +49-511-532-9590 (A.S.); +34-948194700 (ext. 812029) (S.V.)
| | - Silvestre Vicent
- Center for Applied Medical Research, Program in Solid Tumors, University of Navarra, 31008 Pamplona, Spain; (O.E.); (A.V.)
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
- Correspondence: (A.S.); (S.V.); Tel.: +49-511-532-9590 (A.S.); +34-948194700 (ext. 812029) (S.V.)
| |
Collapse
|
20
|
Vicent S, Lieshout R, Saborowski A, Verstegen MMA, Raggi C, Recalcati S, Invernizzi P, van der Laan LJW, Alvaro D, Calvisi DF, Cardinale V. Experimental models to unravel the molecular pathogenesis, cell of origin and stem cell properties of cholangiocarcinoma. Liver Int 2019; 39 Suppl 1:79-97. [PMID: 30851232 DOI: 10.1111/liv.14094] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/10/2019] [Accepted: 02/25/2019] [Indexed: 12/11/2022]
Abstract
Human cholangiocarcinoma (CCA) is an aggressive tumour entity arising from the biliary tree, whose molecular pathogenesis remains largely undeciphered. Over the last decade, the advent of high-throughput and cell-based techniques has significantly increased our knowledge on the molecular mechanisms underlying this disease while, at the same time, unravelling CCA complexity. In particular, it becomes clear that CCA displays pronounced inter- and intratumoural heterogeneity, which is presumably the consequence of the interplay between distinct tissues and cells of origin, the underlying diseases, and the associated molecular alterations. To better characterize these events and to design novel and more effective therapeutic strategies, a number of CCA experimental and preclinical models have been developed and are currently generated. This review summarizes the current knowledge and understanding of these models, critically underlining their translational usefulness and limitations. Furthermore, this review aims to provide a comprehensive overview on cells of origin, cancers stem cells and their dynamic interplay within CCA tissue.
Collapse
Affiliation(s)
- Silvestre Vicent
- Program in Solid Tumors, Center for Applied Applied Medical Research, University of Navarra, Pamplona, Spain.,IdiSNA, Navarra Institute for Health Research, Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Ruby Lieshout
- Department of Surgery, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Anna Saborowski
- Department of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Monique M A Verstegen
- Department of Surgery, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Chiara Raggi
- Humanitas Clinical and Research Center, Rozzano, Italy.,Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Stefania Recalcati
- Department of Biomedical Sciences for Health, University of Milan, Milano, Italy
| | - Pietro Invernizzi
- Division of Gastroenterology and Center of Autoimmune Liver Diseases, Department of Medicine and Surgery, San Gerardo Hospita, l, University of Milano, Bicocca, Italy
| | - Luc J W van der Laan
- Department of Surgery, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Domenico Alvaro
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Diego F Calvisi
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Vincenzo Cardinale
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
21
|
Fouassier L, Marzioni M, Afonso MB, Dooley S, Gaston K, Giannelli G, Rodrigues CMP, Lozano E, Mancarella S, Segatto O, Vaquero J, Marin JJG, Coulouarn C. Signalling networks in cholangiocarcinoma: Molecular pathogenesis, targeted therapies and drug resistance. Liver Int 2019; 39 Suppl 1:43-62. [PMID: 30903728 DOI: 10.1111/liv.14102] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/25/2019] [Accepted: 02/27/2019] [Indexed: 12/13/2022]
Abstract
Cholangiocarcinoma (CCA) is a deadly disease. While surgery may attain cure in a minor fraction of cases, therapeutic options in either the adjuvant or advanced setting are limited. The possibility of advancing the efficacy of therapeutic approaches to CCA relies on understanding its molecular pathogenesis and developing rational therapies aimed at interfering with oncogenic signalling networks that drive and sustain cholangiocarcinogenesis. These efforts are complicated by the intricate biology of CCA, which integrates not only the driving force of tumour cell-intrinsic alterations at the genetic and epigenetic level but also pro-tumorigenic cues conveyed to CCA cells by different cell types present in the rich tumour stroma. Herein, we review our current understanding of the mechanistic bases underpinning the activation of major oncogenic pathways causative of CCA pathogenesis. We subsequently discuss how this knowledge is being exploited to implement rationale-based and genotype-matched therapeutic approaches that predictably will radically transform CCA clinical management in the next decade. We conclude by highlighting the mechanisms of therapeutic resistance in CCA and reviewing innovative approaches to combat resistance at the preclinical and clinical level.
Collapse
Affiliation(s)
- Laura Fouassier
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
| | - Marco Marzioni
- Clinic of Gastroenterology and Hepatology, Università Politecnica delle Marche, Ospedali Riuniti - University Hospital, Ancona, Italy
| | - Marta B Afonso
- Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
| | - Steven Dooley
- Department of Medicine II, Molecular Hepatology Section, Heidelberg University, Mannheim, Germany
| | - Kevin Gaston
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| | - Gianluigi Giannelli
- National Institute of Gastroenterology "Saverio de Bellis", Research Hospital, Bari, Italy
| | - Cecilia M P Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
| | - Elisa Lozano
- Experimental Hepatology and Drug Targeting (HEVEFARM), IBSAL, University of Salamanca, Salamanca, Spain
| | - Serena Mancarella
- National Institute of Gastroenterology "Saverio de Bellis", Research Hospital, Bari, Italy
| | - Oreste Segatto
- Unit of Oncogenomics and Epigenetics, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Javier Vaquero
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.,Sorbonne Université, CNRS, Ecole Polytech., Univ. Paris-Sud, Observatoire de Paris, Université Paris-Saclay, PSL Research University, Paris, France
| | - Jose J G Marin
- Experimental Hepatology and Drug Targeting (HEVEFARM), IBSAL, University of Salamanca, Salamanca, Spain
| | - Cédric Coulouarn
- Inserm, Univ Rennes, Inra, Institut NuMeCan (Nutrition Metabolisms and Cancer), Rennes, France
| |
Collapse
|
22
|
Chen Z, Guo P, Xie X, Yu H, Wang Y, Chen G. The role of tumour microenvironment: a new vision for cholangiocarcinoma. J Cell Mol Med 2018; 23:59-69. [PMID: 30394682 PMCID: PMC6307844 DOI: 10.1111/jcmm.13953] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 09/10/2018] [Indexed: 12/18/2022] Open
Abstract
Cholangiocarcinoma (CCA) is a relatively rare malignant and lethal tumour derived from bile duct epithelium and the morbidity is now increasing worldwide. This disease is difficult to diagnose at its inchoate stage and has poor prognosis. Therefore, a clear understanding of pathogenesis and major influencing factors is the key to develop effective therapeutic methods for CCA. In previous studies, canonical correlation analysis has demonstrated that tumour microenvironment plays an intricate role in the progression of various types of cancers including CCA. CCA tumour microenvironment is a dynamic environment consisting of authoritative tumour stromal cells and extracellular matrix where tumour stromal cells and cancer cells can thrive. CCA stromal cells include immune and non‐immune cells, such as inflammatory cells, endothelial cells, fibroblasts, and macrophages. Likewise, CCA tumour microenvironment contains abundant proliferative factors and can significantly impact the behaviour of cancer cells. Through abominably intricate interactions with CCA cells, CCA tumour microenvironment plays an important role in promoting tumour proliferation, accelerating neovascularization, facilitating tumour invasion, and preventing tumour cells from organismal immune reactions and apoptosis. This review summarizes the recent research progress regarding the connection between tumour behaviours and tumour stromal cells in CCA, as well as the mechanism underlying the effect of tumour stromal cells on the growth of CCA. A thorough understanding of the relationship between CCA and tumour stromal cells can shed some light on the development of new therapeutic methods for treating CCA.
Collapse
Affiliation(s)
- Ziyan Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Pengyi Guo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xiaozai Xie
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Haitao Yu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yi Wang
- Environmental and Public, Health School of Wenzhou Medical University, Wenzhou, China
| | - Gang Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
23
|
Wang J, Dong M, Xu Z, Song X, Zhang S, Qiao Y, Che L, Gordan J, Hu K, Liu Y, Calvisi DF, Chen X. Notch2 controls hepatocyte-derived cholangiocarcinoma formation in mice. Oncogene 2018; 37:3229-3242. [PMID: 29545603 PMCID: PMC6002343 DOI: 10.1038/s41388-018-0188-1] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 12/01/2017] [Accepted: 02/06/2018] [Indexed: 02/08/2023]
Abstract
Liver cancer comprises a group of malignant tumors, among which hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC) are the most common. ICC is especially pernicious and associated with poor clinical outcome. Studies have shown that a subset of human ICCs may originate from mature hepatocytes. However, the mechanisms driving the trans-differentiation of hepatocytes into malignant cholangiocytes remain poorly defined. We adopted lineage tracing techniques and an established murine hepatocyte-derived ICC model by hydrodynamic injection of activated forms of AKT (myr-AKT) and Yap (YapS127A) proto-oncogenes. Wild-type, Notch1 flox/flox , and Notch2 flox/flox mice were used to investigate the role of canonical Notch signaling and Notch receptors in AKT/Yap-driven ICC formation. Human ICC and HCC cell lines were transfected with siRNA against Notch2 to determine whether Notch2 regulates biliary marker expression in liver tumor cells. We found that AKT/Yap-induced ICC formation is hepatocyte derived and this process is strictly dependent on the canonical Notch signaling pathway in vivo. Deletion of Notch2 in AKT/Yap-induced tumors switched the phenotype from ICC to hepatocellular adenoma-like lesions, while inactivation of Notch1 in hepatocytes did not result in significant histomorphological changes. Finally, in vitro studies revealed that Notch2 silencing in ICC and HCC cell lines down-regulates the expression of Sox9 and EpCAM biliary markers. Notch2 is the major determinant of hepatocyte-derived ICC formation in mice.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- Bile Duct Neoplasms/metabolism
- Bile Duct Neoplasms/pathology
- Bile Ducts, Intrahepatic/pathology
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Cell Cycle Proteins
- Cell Line, Tumor
- Cholangiocarcinoma/metabolism
- Cholangiocarcinoma/pathology
- Female
- Hepatocytes/metabolism
- Hepatocytes/pathology
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Male
- Mice, Transgenic
- Phosphoproteins/genetics
- Phosphoproteins/metabolism
- Proto-Oncogene Proteins c-akt/metabolism
- Receptor, Notch1/genetics
- Receptor, Notch1/metabolism
- Receptor, Notch2/genetics
- Receptor, Notch2/metabolism
- Signal Transduction/physiology
- YAP-Signaling Proteins
Collapse
Affiliation(s)
- Jingxiao Wang
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, USA
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Mingjie Dong
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, USA
- 307 Hospital of Academy of Military Medical Science, Beijing, China
| | - Zhong Xu
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, USA
- Department of Gastroenterology, Guizhou Provincial People's Hospital, Guizhou, China
| | - Xinhua Song
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, USA
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Shanshan Zhang
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, USA
| | - Yu Qiao
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, USA
- Department of Oncology, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Li Che
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, USA
| | - John Gordan
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, USA
| | - Kaiwen Hu
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yan Liu
- 307 Hospital of Academy of Military Medical Science, Beijing, China.
| | - Diego F Calvisi
- Institut für Pathologie, Universitätsmedizin Greifswald, Greifswald, Germany.
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, USA.
| |
Collapse
|
24
|
Luo H, Liu WH, Liang HY, Yan HT, Lin N, Li DY, Wang T, Tang LJ. Differentiation-inducing therapeutic effect of Notch inhibition in reversing malignant transformation of liver normal stem cells via MET. Oncotarget 2018; 9:18885-18895. [PMID: 29721169 PMCID: PMC5922363 DOI: 10.18632/oncotarget.24421] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 01/01/2018] [Indexed: 12/27/2022] Open
Abstract
Background Liver cancer stem cells (LCSCs) are the key factors for cancer metastasis, recurrent, and drug resistance. LCSCs are originated from either hepatocytes dedifferentiation or differentiation arresting of liver normal stem cells (LNSCs). Differentiation-inducing therapy is a novel strategy in solid tumors. Furthermore, Notch signaling pathway has been proved to play important role in the process of hepatocytes differentiation. In previous study, a malignant transformation cellular model of LNSCs has been built up, and in this study we are trying to illustrate whether inhibition of Notch can reverse this malignant tendency and drive these malignant cells back to differentiate into mature hepatocytes. Results Inhibition of Notch signaling pathway can down-regulate the stemness-related cancer markers, lower the proliferative status, alleviate the invasive characteristic, or attenuate the metastasis tendency. What is more, it can help the malignantly transformed cells to regain the mature hepatic function of glucagon synthesis, urea metabolism, albumin production, and indocyanine-green (ICG) clearance. Materials and Methods HOX transcript antisense RNA (HOTAIR) expression was enhanced in LNSCs via lentivirus transduction to set up the malignant transformation cellular model. Then, a Notch inhibitor was applied to induce malignantly transformed cells differentiate into mature hepatocytes, and malignant abilities of proliferation, invasiveness, tumorigenesis as well as mature hepatocyte function were observed and compared. Conclusions The data demonstrate that the anti-tumor effects of Notch inhibition may lie not only on killing the cancer cells or LCSCs directly, it can also induce the LCSCs differentiation into mature hepatocytes via mesenchymal-epithelial transition (MET) progress or downgrade the malignancy.
Collapse
Affiliation(s)
- Hao Luo
- Third Military Medical University, Chongqing 400038, China.,General Surgery Center, Chengdu Military General Hospital, Chengdu 610083, China
| | - Wei-Hui Liu
- General Surgery Center, Chengdu Military General Hospital, Chengdu 610083, China
| | - Hong-Yin Liang
- General Surgery Center, Chengdu Military General Hospital, Chengdu 610083, China
| | - Hong-Tao Yan
- General Surgery Center, Chengdu Military General Hospital, Chengdu 610083, China
| | - Ning Lin
- Department of Clinical Nutrition, Chengdu Military General Hospital, Chengdu 610083, China
| | - Dong-Yu Li
- General Surgery Center, Chengdu Military General Hospital, Chengdu 610083, China
| | - Tao Wang
- General Surgery Center, Chengdu Military General Hospital, Chengdu 610083, China
| | - Li-Jun Tang
- Third Military Medical University, Chongqing 400038, China.,General Surgery Center, Chengdu Military General Hospital, Chengdu 610083, China
| |
Collapse
|
25
|
Zhang S, Wang J, Wang H, Fan L, Fan B, Zeng B, Tao J, Li X, Che L, Cigliano A, Ribback S, Dombrowski F, Chen B, Cong W, Wei L, Calvisi DF, Chen X. Hippo Cascade Controls Lineage Commitment of Liver Tumors in Mice and Humans. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:995-1006. [PMID: 29378174 DOI: 10.1016/j.ajpath.2017.12.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 12/11/2017] [Accepted: 12/28/2017] [Indexed: 02/05/2023]
Abstract
Primary liver cancer consists mainly of hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC). A subset of human HCCs expresses a ICC-like gene signature and is classified as ICC-like HCC. The Hippo pathway is a critical regulator of normal and malignant liver development. However, the precise function(s) of the Hippo cascade along liver carcinogenesis remain to be fully delineated. The role of the Hippo pathway in a murine mixed HCC/ICC model induced by activated forms of AKT and Ras oncogenes (AKT/Ras) was investigated. The authors demonstrated the inactivation of Hippo in AKT/Ras liver tumors leading to nuclear localization of Yap and TAZ. Coexpression of AKT/Ras with Lats2, which activates Hippo, or the dominant negative form of TEAD2 (dnTEAD2), which blocks Yap/TAZ activity, resulted in delayed hepatocarcinogenesis and elimination of ICC-like lesions in the liver. Mechanistically, Notch2 expression was found to be down-regulated by the Hippo pathway in liver tumors. Overexpression of Lats2 or dnTEAD2 in human HCC cell lines inhibited their growth and led to the decreased expression of ICC-like markers, as well as Notch2 expression. Altogether, this study supports the key role of the Hippo cascade in regulating the differentiation status of liver tumors.
Collapse
Affiliation(s)
- Shanshan Zhang
- Tumor Immunology and Gene Therapy Center, Second Military Medical University, Shanghai, China; Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California; Department of Pathology, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Jingxiao Wang
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California; Second Clinical Medical School, Beijing University of Chinese Medicine, Beijing, China
| | - Haichuan Wang
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California; Liver Transplantation Division, Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Lingling Fan
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California
| | - Biao Fan
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California; Department of Gastrointestinal Surgery, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing, China
| | - Billy Zeng
- Department of Pediatrics, University of California, San Francisco, California; Institute for Computational Health Sciences, University of California, San Francisco, California
| | - Junyan Tao
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California
| | - Xiaolei Li
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California
| | - Li Che
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California
| | - Antonio Cigliano
- National Institute of Gastroenterology "S. de Bellis", Research Hospital, Castellana Grotte, Italy
| | - Silvia Ribback
- Institute of Pathology, University of Greifswald, Greifswald, Germany
| | - Frank Dombrowski
- Institute of Pathology, University of Greifswald, Greifswald, Germany
| | - Bin Chen
- Department of Pediatrics, University of California, San Francisco, California; Institute for Computational Health Sciences, University of California, San Francisco, California
| | - Wenming Cong
- Department of Pathology, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Lixin Wei
- Tumor Immunology and Gene Therapy Center, Second Military Medical University, Shanghai, China
| | - Diego F Calvisi
- Institute of Pathology, University of Greifswald, Greifswald, Germany.
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California.
| |
Collapse
|
26
|
Matson DR, Hardin H, Buehler D, Lloyd RV. AKT activity is elevated in aggressive thyroid neoplasms where it promotes proliferation and invasion. Exp Mol Pathol 2017; 103:288-293. [PMID: 29169802 DOI: 10.1016/j.yexmp.2017.11.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 11/18/2017] [Indexed: 02/06/2023]
Abstract
The PI3K/AKT/mTOR signaling pathway controls major cellular processes such as cell growth, proliferation and survival. Stimulation of this pathway leads to AKT phosphorylation and activation, resulting in phosphorylation of mTOR and myriad other targets. AKT upregulation has been implicated in thyroid cancer pathogenesis and is a candidate treatment target for patients with advanced disease that has not responded to traditional therapies. Here we evaluate a large series of benign and malignant thyroid tumors for AKT activity and intracellular distribution. We also deplete AKT from multiple thyroid cancer cell lines, including putative cancer stem cell lines, and measure the effect on proliferation and invasion in vitro. We show that active AKT has a predominantly nuclear distribution and its expression is highest in anaplastic thyroid carcinomas and papillary thyroid carcinomas, including encapsulated and invasive follicular variants. Depletion of AKT in thyroid carcinoma cell lines led to greatly reduced proliferative capacity and resulted in a reduction of invasive potential. A reduction in invasion was also observed in the cancer stem cell compartment. Targeting AKT activity in the clinical setting may slow the growth and spread of aggressive thyroid neoplasms, and target the tumor stem cell compartment.
Collapse
Affiliation(s)
- Daniel R Matson
- Department of Pathology and Laboratory Medicine, University of Wisconsin, School of Medicine and Public Health, 600 Highland Ave, Madison, WI 53792, USA
| | - Heather Hardin
- Department of Pathology and Laboratory Medicine, University of Wisconsin, School of Medicine and Public Health, 600 Highland Ave, Madison, WI 53792, USA
| | - Darya Buehler
- Department of Pathology and Laboratory Medicine, University of Wisconsin, School of Medicine and Public Health, 600 Highland Ave, Madison, WI 53792, USA
| | - Ricardo V Lloyd
- Department of Pathology and Laboratory Medicine, University of Wisconsin, School of Medicine and Public Health, 600 Highland Ave, Madison, WI 53792, USA.
| |
Collapse
|
27
|
Cigliano A, Wang J, Chen X, Calvisi DF. Role of the Notch signaling in cholangiocarcinoma. Expert Opin Ther Targets 2017; 21:471-483. [PMID: 28326864 DOI: 10.1080/14728222.2017.1310842] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Cholangiocarcinoma (CCA) is an emerging cancer entity of the liver, associated with poor outcome and characterized by resistance to conventional chemotherapeutic treatments. In the last decade, many signaling pathways associated with CCA development and progression have been identified and are currently under intense investigation. Cumulating evidence indicates that the Notch cascade, a highly-conserved pathway in most multicellular organisms, is a critical player both in liver malignant transformation and tumor aggressiveness, thus representing a potential therapeutic target in this pernicious disease. Areas covered: In the present review article, we comprehensively summarize and critically discuss the current knowledge on the Notch pathway, its specific and key roles in cholangiocarcinogenesis, the treatment strategies aimed at suppressing this signaling cascade in cancer, and the encouraging results coming from preclinical trials. Expert opinion: The Notch pathway represents a major driver of carcinogenesis and a promising therapeutic target in human CCA. A better understanding of the molecular mechanisms triggered by the Notch pathway as well as its functional crosstalk with other signaling cascade will be highly helpful for the design of innovative therapies against human CCA.
Collapse
Affiliation(s)
- Antonio Cigliano
- a Institut für Pathologie , Universitätsmedizin Greifswald , Greifswald , Germany
| | - Jingxiao Wang
- b Second Clinical Medical School , Beijing University of Chinese Medicine , Beijing , China.,c Department of Bioengineering and Therapeutic Sciences and Liver Center , University of California , San Francisco , CA , USA
| | - Xin Chen
- c Department of Bioengineering and Therapeutic Sciences and Liver Center , University of California , San Francisco , CA , USA
| | - Diego F Calvisi
- a Institut für Pathologie , Universitätsmedizin Greifswald , Greifswald , Germany
| |
Collapse
|