1
|
Liu Z, Zeng Y, Li R, Yan Y, Yi S, Zhang K. Treatment of chronic obstructive pulmonary disease by traditional Chinese medicine Morin monomer regulated by autophagy. J Thorac Dis 2024; 16:6052-6063. [PMID: 39444855 PMCID: PMC11494543 DOI: 10.21037/jtd-23-1836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 08/06/2024] [Indexed: 10/25/2024]
Abstract
Background Chronic obstructive pulmonary disease (COPD) is a frequently occurring disorder. The aim of this study is to explore the mechanism of traditional Chinese medicine Morin monomer in the treatment of COPD via regulating autophagy based on the long non-coding RNA (lncRNA) H19/microRNA (miR)-194-5p/Sirtuin (SIRT)1 signal axis. Methods The COPD rat model was constructed, and the lung tissues were collected. The pathological analysis was performed using hematoxylin-eosin (HE), Masson, and periodic acid-Schiff (PAS) staining. Autophagosomes were observed using transmission electron microscope. LncRNA H19, miR-194-5p, SIRT1 genes in the rat lung tissues were detected using reverse transcription-quantitative real-time polymerase chain reaction (RT-qPCR). The autophagy-related proteins including SIRT1, mammalian/mechanistic target of rapamycin (mTOR), phosphorylated (p)-mTOR, microtubule-associated protein light chain 3 (LC3), Beclin-1, autophagy-related (ATG)7, and p62 in each group were detected using Western blot. Results The rats in the control group had normal lung structure. Alveolar enlargement and destruction could be found in the rat lung tissues in the model group, accompanied with obvious infiltration of inflammatory cells, thickened bronchial walls, enlarged alveolar septum, collagen fibers deposition, and goblet cells proliferation. In comparison with the model group, Morin treatment relieved the lung injuries, which was optimized in the moderate- and high-dose groups. The number of autophagosomes in the lung tissues of the model rats was dramatically increased compared with the normal rats. However, the number of autophagosomes in each Morin treatment group was obviously less than that in the model group. LncRNA H19 and SIRT1 expression was significantly increased in the model group, and miR-194-5p was significantly decreased (P<0.05). Morin and 3-methyladenine (3-MA) could obviously reduce the lncRNA H19 and SIRT1 expression, and increase the miR-194-5p expression (P<0.05). Relative to control rats, ATG7, Beclin-1, LC3II/I and SIRT1 levels in the model group increased obviously, while the expression of p62, and p-mTOR/mTOR decreased (P<0.05). Morin treatment reduced the expression of ATG7, Beclin-1, SIRT1, LC3II/I significantly, and increased the p-mTOR/mTOR and p62 expression (P<0.05). Conclusions Morin decreased lncRNA H19 expression, resulting in upregulation of miR-194-5p expression, downregulation of SIRT1 expression, and increased of p-mTOR/mTOR expression. Furthermore, cell autophagy was inhibited, contributing to the COPD treatment.
Collapse
Affiliation(s)
- Zhen Liu
- Department of Traditional Chinese Medicine, Guizhou Provincial People's Hospital, Guiyang, China
| | - Yang Zeng
- Department of Traditional Chinese Medicine, Guizhou Provincial People's Hospital, Guiyang, China
| | - Rui Li
- Department of Traditional Chinese Medicine, Guizhou Provincial People's Hospital, Guiyang, China
| | - Ying Yan
- Department of Traditional Chinese Medicine, Guizhou Provincial People's Hospital, Guiyang, China
| | - Sicheng Yi
- Department of Traditional Chinese Medicine, Guizhou Provincial People's Hospital, Guiyang, China
| | - Kui Zhang
- Department of Traditional Chinese Medicine, Guizhou Provincial People's Hospital, Guiyang, China
| |
Collapse
|
2
|
Panettieri E, Campisi A, De Rose AM, Mele C, Giuliante F, Vauthey JN, Ardito F. Emerging Prognostic Markers in Patients Undergoing Liver Resection for Hepatocellular Carcinoma: A Narrative Review. Cancers (Basel) 2024; 16:2183. [PMID: 38927889 PMCID: PMC11201456 DOI: 10.3390/cancers16122183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/27/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
In patients with hepatocellular carcinoma (HCC), liver resection is potentially curative. Nevertheless, post-operative recurrence is common, occurring in up to 70% of patients. Factors traditionally recognized to predict recurrence and survival after liver resection for HCC include pathologic factors (i.e., microvascular and capsular invasion) and an increase in alpha-fetoprotein level. During the past decade, many new markers have been reported to correlate with prognosis after resection of HCC: liquid biopsy markers, gene signatures, inflammation markers, and other biomarkers, including PIVKA-II, immune checkpoint molecules, and proteins in urinary exosomes. However, not all of these new markers are readily available in clinical practice, and their reproducibility is unclear. Liquid biopsy is a powerful and established tool for predicting long-term outcomes after resection of HCC; the main limitation of liquid biopsy is represented by the cost related to its technical implementation. Numerous patterns of genetic expression capable of predicting survival after curative-intent hepatectomy for HCC have been identified, but published findings regarding these markers are heterogenous. Inflammation markers in the form of prognostic nutritional index and different blood cell ratios seem more easily reproducible and more affordable on a large scale than other emerging markers. To select the most effective treatment for patients with HCC, it is crucial that the scientific community validate new predictive markers for recurrence and survival after resection that are reliable and widely reproducible. More reports from Western countries are necessary to corroborate the evidence.
Collapse
Affiliation(s)
- Elena Panettieri
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Andrea Campisi
- Hepatobiliary Surgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (A.C.); (A.M.D.R.); (C.M.); (F.G.); (F.A.)
| | - Agostino M. De Rose
- Hepatobiliary Surgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (A.C.); (A.M.D.R.); (C.M.); (F.G.); (F.A.)
| | - Caterina Mele
- Hepatobiliary Surgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (A.C.); (A.M.D.R.); (C.M.); (F.G.); (F.A.)
| | - Felice Giuliante
- Hepatobiliary Surgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (A.C.); (A.M.D.R.); (C.M.); (F.G.); (F.A.)
| | - Jean-Nicolas Vauthey
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Francesco Ardito
- Hepatobiliary Surgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (A.C.); (A.M.D.R.); (C.M.); (F.G.); (F.A.)
| |
Collapse
|
3
|
Apostolo D, Ferreira LL, Vincenzi F, Vercellino N, Minisini R, Latini F, Ferrari B, Burlone ME, Pirisi M, Bellan M. From MASH to HCC: the role of Gas6/TAM receptors. Front Immunol 2024; 15:1332818. [PMID: 38298195 PMCID: PMC10827955 DOI: 10.3389/fimmu.2024.1332818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 01/02/2024] [Indexed: 02/02/2024] Open
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is the replacement term for what used to be called nonalcoholic steatohepatitis (NASH). It is characterized by inflammation and injury of the liver in the presence of cardiometabolic risk factors and may eventually result in the development of hepatocellular carcinoma (HCC), the most common form of primary liver cancer. Several pathogenic mechanisms are involved in the transition from MASH to HCC, encompassing metabolic injury, inflammation, immune dysregulation and fibrosis. In this context, Gas6 (Growth Arrest-Specific 6) and TAM (Tyro3, Axl, and MerTK) receptors may play important roles. The Gas6/TAM family is involved in the modulation of inflammation, lipid metabolism, fibrosis, tumor progression and metastasis, processes which play an important role in the pathophysiology of acute and chronic liver diseases. In this review, we discuss MASH-associated HCC and the potential involvement of the Gas6/TAM system in disease development and progression. In addition, since therapeutic strategies for MASH and HCC are limited, we also speculate regarding possible future treatments involving the targeting of Gas6 or TAM receptors.
Collapse
Affiliation(s)
- Daria Apostolo
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Luciana L. Ferreira
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Federica Vincenzi
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Nicole Vercellino
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Rosalba Minisini
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Federico Latini
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Barbara Ferrari
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Michela E. Burlone
- Department of Internal Medicine, Azienda Ospedaliero-Universitaria Maggiore Della Carità, Novara, Italy
| | - Mario Pirisi
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
- Department of Internal Medicine, Azienda Ospedaliero-Universitaria Maggiore Della Carità, Novara, Italy
- Center on Autoimmune and Allergic Diseases, Università del Piemonte Orientale, Novara, Italy
| | - Mattia Bellan
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
- Department of Internal Medicine, Azienda Ospedaliero-Universitaria Maggiore Della Carità, Novara, Italy
- Center on Autoimmune and Allergic Diseases, Università del Piemonte Orientale, Novara, Italy
| |
Collapse
|
4
|
Hayashi M, Abe K, Sugaya T, Takahata Y, Fujita M, Takahashi A, Ohira H. Influence of serum Gas6 levels on prognosis in patients with hepatocellular carcinoma. Jpn J Clin Oncol 2024; 54:62-69. [PMID: 37801445 DOI: 10.1093/jjco/hyad132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 09/15/2023] [Indexed: 10/08/2023] Open
Abstract
OBJECTIVE The prediction of prognosis in hepatocellular carcinoma patients is important for switching treatment. The association between circulating growth arrest-specific 6 levels and prognosis in hepatocellular carcinoma patients is unknown. METHODS We retrospectively analysed the association between serum growth arrest-specific 6 levels and clinical findings in 132 patients with hepatocellular carcinoma. Serum growth arrest-specific 6 levels were measured using enzyme-linked immunosorbent assay. RESULTS Amongst 132 patients, the Barcelona Clinic Liver Cancer stage was classified as 0, A, B, C and D in 19, 48, 41, 18 and 6 patients, respectively. Serum growth arrest-specific 6 levels in hepatocellular carcinoma patients were higher than those in healthy controls (28.4 ng/mL vs. 19.6 ng/mL, P < 0.001), and growth arrest-specific 6 levels were positively correlated with soluble Axl levels. In the entire cohort, high growth arrest-specific 6 levels were associated with a shorter survival period (hazard ratio: 1.78 per 20 ng/mL, 95% confidence interval: 1.01-3.16, P = 0.045). In early and intermediate-stage hepatocellular carcinoma patients treated with transcatheter arterial chemoembolization (n = 59), we determined a cut-off value of 36.4 ng/mL based on the receiver operating characteristic curve to predict death within 3 years, and high growth arrest-specific 6 levels were associated with a high cumulative incidence of portal vein tumour thrombosis (Gray's test: P = 0.010) and shorter overall survival (log-rank: P = 0.005). CONCLUSIONS Serum growth arrest-specific 6 levels were associated with prognosis in hepatocellular carcinoma patients. In early and intermediate-stage hepatocellular carcinoma patients who underwent transcatheter arterial chemoembolization, high growth arrest-specific 6 levels were associated with a high incidence of portal vein tumour thrombosis. Circulating growth arrest-specific 6 levels may be a useful prognostic marker in hepatocellular carcinoma patients.
Collapse
Affiliation(s)
- Manabu Hayashi
- Department of Gastroenterology, Fukushima Medical University, Fukushima, Japan
| | - Kazumichi Abe
- Department of Gastroenterology, Fukushima Medical University, Fukushima, Japan
| | - Tatsuro Sugaya
- Department of Gastroenterology, Fukushima Medical University, Fukushima, Japan
| | - Yosuke Takahata
- Department of Gastroenterology, Fukushima Medical University, Fukushima, Japan
| | - Masashi Fujita
- Department of Gastroenterology, Fukushima Medical University, Fukushima, Japan
| | - Atsushi Takahashi
- Department of Gastroenterology, Fukushima Medical University, Fukushima, Japan
| | - Hiromasa Ohira
- Department of Gastroenterology, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
5
|
Tang Y, Zang H, Wen Q, Fan S. AXL in cancer: a modulator of drug resistance and therapeutic target. J Exp Clin Cancer Res 2023; 42:148. [PMID: 37328828 DOI: 10.1186/s13046-023-02726-w] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 05/31/2023] [Indexed: 06/18/2023] Open
Abstract
AXL is a member of the TAM (TYRO3, AXL, and MERTK) receptor tyrosine kinases family (RTKs), and its abnormal expression has been linked to clinicopathological features and poor prognosis of cancer patients. There is mounting evidence supporting AXL's role in the occurrence and progression of cancer, as well as drug resistance and treatment tolerance. Recent studies revealed that reducing AXL expression can weaken cancer cells' drug resistance, indicating that AXL may be a promising target for anti-cancer drug treatment. This review aims to summarize the AXL's structure, the mechanisms regulating and activating it, and its expression pattern, especially in drug-resistant cancers. Additionally, we will discuss the diverse functions of AXL in mediating cancer drug resistance and the potential of AXL inhibitors in cancer treatment.
Collapse
Affiliation(s)
- Yaoxiang Tang
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Hongjing Zang
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Qiuyuan Wen
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| | - Songqing Fan
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
6
|
Belyaeva E, Loginova N, Schroeder BA, Goldlust IS, Acharya A, Kumar S, Timashev P, Ulasov I. The spectrum of cell death in sarcoma. Biomed Pharmacother 2023; 162:114683. [PMID: 37031493 DOI: 10.1016/j.biopha.2023.114683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/01/2023] [Accepted: 04/06/2023] [Indexed: 04/11/2023] Open
Abstract
The balance between cell death and cell survival is a highly coordinated process by which cells break down and remove unnecessary or harmful materials in a controlled, highly regulated, and compartmentalized manner. Cell exposure to various stresses, such as oxygen starvation, a lack of nutrients, or exposure to radiation, can initiate autophagy. Autophagy is a carefully orchestrated process with multiple steps, each regulated by specific genes and proteins. Autophagy proteins impact cellular maintenance and cell fate in response to stress, and targeting this process is one of the most promising methods of anti-tumor therapy. It is currently not fully understood how autophagy affects different types of tumor cells, which makes it challenging to predict outcomes when this process is manipulated. In this review, we will explore the mechanisms of autophagy and investigate it as a potential and promising therapeutic target for aggressive sarcomas.
Collapse
Affiliation(s)
- Elizaveta Belyaeva
- Group of Experimental Biotherapy and Diagnostics, Institute for Regenerative Medicine, World-Class Research Centre "Digital Biodesign and Personalized Healthcare", I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Nina Loginova
- Group of Experimental Biotherapy and Diagnostics, Institute for Regenerative Medicine, World-Class Research Centre "Digital Biodesign and Personalized Healthcare", I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Brett A Schroeder
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20814, USA
| | - Ian S Goldlust
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20814, USA
| | - Arbind Acharya
- Laboratory of Cancer Immunology, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India
| | - Sandeep Kumar
- Laboratory of Cancer Immunology, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India
| | - Peter Timashev
- World-Class Research Centre "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Ilya Ulasov
- Group of Experimental Biotherapy and Diagnostics, Institute for Regenerative Medicine, World-Class Research Centre "Digital Biodesign and Personalized Healthcare", I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russia.
| |
Collapse
|
7
|
Wang Y, Tsai M, Chen Y, Hsieh P, Hung C, Lin H, Hsu Y, Yeh J, Hsiao P, Su Y, Ma C, Lee C, Lin C, Shu C, Li Y, Tsai M, Lin JY, Peng W, Yu M, Lin C. NPRL2 down-regulation facilitates the growth of hepatocellular carcinoma via the mTOR pathway and autophagy suppression. Hepatol Commun 2022; 6:3563-3577. [PMID: 36321403 PMCID: PMC9701468 DOI: 10.1002/hep4.2019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 04/27/2022] [Accepted: 05/17/2022] [Indexed: 11/28/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a highly invasive malignancy. Recently, GATOR1 (Gap Activity TOward Rags 1) complexes have been shown to play an important role in regulating tumor growth. NPRL2 is a critical component of the GATOR1 complex. Therefore, this study used NPRL2 knockdown to investigate how GATORC1 regulates the prognosis and development of HCC via the mammalian target of rapamycin (mTOR) and autophagy signaling pathways. We established HepG2 cells with NPRL2 knockdown using small interfering RNA (siRNA) and short hairpin RNA (shRNA) systems. The siRNA-mediated and shRNA-mediated NPRL2 down-regulation significantly reduced the expression of NPRL2 and two other GATPOR1 complex components, NPRL3 and DEPDC5, in HepG2 cells; furthermore, the efficient down-regulation of NPRL2 protein expression by both the shRNA and siRNA systems enhanced the proliferation, migration, and colony formation in vitro. Additionally, the NPRL2 down-regulation significantly increased HCC growth in the subcutaneous and orthotopic xenograft mouse models. The NPRL2 down-regulation increased the Rag GTPases and mTOR activation and inhibited autophagy in vitro and in vivo. Moreover, the NPRL2 level in the tumors was significantly associated with mortality, recurrence, the serum alpha fetoprotein level, the tumor size, the American Joint Committee on Cancer stage, and the Barcelona Clinic Liver Cancer stage. Low NPRL2, NPRL3, DEPDC5, and LC3, and high p62 and mTOR protein expression in the tumors was significantly associated with disease-free survival and overall survival in 300 patients with HCC after surgical resection. Conclusion: The efficient down-regulation of NPRL2 significantly increased HCC proliferation, migration, and colony formation in vitro, and increased HCC growth in vivo. Low NPRL2 protein expression in the tumors was closely correlated with poorer clinical outcomes in patients with HCC. These results provide a mechanistic understanding of HCC and aid the development of treatments for HCC.
Collapse
Affiliation(s)
- Ya‐Chin Wang
- Division of Gastroenterology and HepatologyE‐Da Dachang HospitalI‐Shou UniversityKaohsiungTaiwan,School of MedicineCollege of MedicineI‐Shou UniversityKaohsiungTaiwan
| | - Ming‐Chao Tsai
- Division of Hepato‐GastroenterologyDepartment of MedicineKaohsiung Chang Gung Memorial HospitalChang Gung University College of MedicineKaohsiungTaiwan
| | - Yaw‐Sen Chen
- School of MedicineCollege of MedicineI‐Shou UniversityKaohsiungTaiwan,Department of SurgeryE‐Da HospitalI‐Shou UniversityKaohsiungTaiwan
| | - Pei‐Min Hsieh
- School of MedicineCollege of MedicineI‐Shou UniversityKaohsiungTaiwan,Department of SurgeryE‐Da HospitalI‐Shou UniversityKaohsiungTaiwan
| | - Chao‐Ming Hung
- School of MedicineCollege of MedicineI‐Shou UniversityKaohsiungTaiwan,Department of SurgeryE‐Da HospitalI‐Shou UniversityKaohsiungTaiwan,Department of SurgeryE‐Da Cancer HospitalI‐Shou UniversityKaohsiungTaiwan
| | - Hung‐Yu Lin
- School of MedicineCollege of MedicineI‐Shou UniversityKaohsiungTaiwan,Department of SurgeryE‐Da HospitalI‐Shou UniversityKaohsiungTaiwan,Department of SurgeryE‐Da Cancer HospitalI‐Shou UniversityKaohsiungTaiwan
| | - Yao‐Chun Hsu
- School of MedicineCollege of MedicineI‐Shou UniversityKaohsiungTaiwan,Division of Gastroenterology and HepatologyDepartment of Internal MedicineE‐Da HospitalI‐Shou UniversityKaohsiungTaiwan
| | - Jen‐Hao Yeh
- Division of Gastroenterology and HepatologyE‐Da Dachang HospitalI‐Shou UniversityKaohsiungTaiwan,School of MedicineCollege of MedicineI‐Shou UniversityKaohsiungTaiwan,Division of Gastroenterology and HepatologyDepartment of Internal MedicineE‐Da HospitalI‐Shou UniversityKaohsiungTaiwan
| | - Pojen Hsiao
- Division of Gastroenterology and HepatologyE‐Da Dachang HospitalI‐Shou UniversityKaohsiungTaiwan,School of MedicineCollege of MedicineI‐Shou UniversityKaohsiungTaiwan
| | - Yu‐Cheih Su
- School of MedicineCollege of MedicineI‐Shou UniversityKaohsiungTaiwan,Division of Hematology‐OncologyDepartment of MedicineE‐Da HospitalI‐Shou UniversityKaohsiungTaiwan
| | - Ching‐Hou Ma
- Department of Orthopedic SurgeryE‐Da HospitalI‐Shou UniversityKaohsiungTaiwan
| | - Chih‐Yuan Lee
- Department of SurgeryNational Taiwan University HospitalTaipeiTaiwan
| | - Chih‐Che Lin
- Department of SurgeryKaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiungTaiwan
| | - Chih‐Wen Shu
- School of MedicineCollege of MedicineI‐Shou UniversityKaohsiungTaiwan
| | - Yu‐Chan Li
- Division of Gastroenterology and HepatologyE‐Da Dachang HospitalI‐Shou UniversityKaohsiungTaiwan,School of MedicineCollege of MedicineI‐Shou UniversityKaohsiungTaiwan
| | - Mei‐Hsing Tsai
- School of MedicineCollege of MedicineI‐Shou UniversityKaohsiungTaiwan
| | - James Yu Lin
- Division of Gastroenterology and HepatologyE‐Da Dachang HospitalI‐Shou UniversityKaohsiungTaiwan,School of MedicineCollege of MedicineI‐Shou UniversityKaohsiungTaiwan,Kaohsiung American SchoolKaohsiungTaiwan
| | - Wei‐Hao Peng
- School of MedicineCollege of MedicineI‐Shou UniversityKaohsiungTaiwan
| | - Ming‐Lung Yu
- Hepatobiliary SectionDepartment of Internal Medicine, Hepatitis CenterKaohsiung Medical University HospitalKaohsiungTaiwan,School of Medicine and Hepatitis Research CenterCollege of Medicine and Center for Liquid Biopsy and Cohort ResearchKaohsiung Medical UniversityKaohsiungTaiwan
| | - Chih‐Wen Lin
- Division of Gastroenterology and HepatologyE‐Da Dachang HospitalI‐Shou UniversityKaohsiungTaiwan,School of MedicineCollege of MedicineI‐Shou UniversityKaohsiungTaiwan,Division of Gastroenterology and HepatologyDepartment of Internal MedicineE‐Da HospitalI‐Shou UniversityKaohsiungTaiwan,School of Chinese MedicineCollege of Chinese Medicine, Research Center for Traditional Chinese Medicine China Medical UniversityTaichungTaiwan
| |
Collapse
|
8
|
Engelsen AST, Lotsberg ML, Abou Khouzam R, Thiery JP, Lorens JB, Chouaib S, Terry S. Dissecting the Role of AXL in Cancer Immune Escape and Resistance to Immune Checkpoint Inhibition. Front Immunol 2022; 13:869676. [PMID: 35572601 PMCID: PMC9092944 DOI: 10.3389/fimmu.2022.869676] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/16/2022] [Indexed: 12/12/2022] Open
Abstract
The development and implementation of Immune Checkpoint Inhibitors (ICI) in clinical oncology have significantly improved the survival of a subset of cancer patients with metastatic disease previously considered uniformly lethal. However, the low response rates and the low number of patients with durable clinical responses remain major concerns and underscore the limited understanding of mechanisms regulating anti-tumor immunity and tumor immune resistance. There is an urgent unmet need for novel approaches to enhance the efficacy of ICI in the clinic, and for predictive tools that can accurately predict ICI responders based on the composition of their tumor microenvironment. The receptor tyrosine kinase (RTK) AXL has been associated with poor prognosis in numerous malignancies and the emergence of therapy resistance. AXL is a member of the TYRO3-AXL-MERTK (TAM) kinase family. Upon binding to its ligand GAS6, AXL regulates cell signaling cascades and cellular communication between various components of the tumor microenvironment, including cancer cells, endothelial cells, and immune cells. Converging evidence points to AXL as an attractive molecular target to overcome therapy resistance and immunosuppression, supported by the potential of AXL inhibitors to improve ICI efficacy. Here, we review the current literature on the prominent role of AXL in regulating cancer progression, with particular attention to its effects on anti-tumor immune response and resistance to ICI. We discuss future directions with the aim to understand better the complex role of AXL and TAM receptors in cancer and the potential value of this knowledge and targeted inhibition for the benefit of cancer patients.
Collapse
Affiliation(s)
- Agnete S. T. Engelsen
- Centre for Cancer Biomarkers and Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Maria L. Lotsberg
- Centre for Cancer Biomarkers and Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Raefa Abou Khouzam
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - Jean-Paul Thiery
- Centre for Cancer Biomarkers and Department of Biomedicine, University of Bergen, Bergen, Norway
- Guangzhou Laboratory, Guangzhou, China
- Inserm, UMR 1186, Integrative Tumor Immunology and Immunotherapy, Villejuif, France
| | - James B. Lorens
- Centre for Cancer Biomarkers and Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Salem Chouaib
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
- Inserm, UMR 1186, Integrative Tumor Immunology and Immunotherapy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Faculty of Medicine, University Paris Sud, Le Kremlin-Bicêtre, France
| | - Stéphane Terry
- Inserm, UMR 1186, Integrative Tumor Immunology and Immunotherapy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Faculty of Medicine, University Paris Sud, Le Kremlin-Bicêtre, France
- Research Department, Inovarion, Paris, France
| |
Collapse
|
9
|
Zhang X, Zhou C, Wu F, Gao C, Liu Q, Lv P, Li M, Huang L, Wu T, Li W. Bio-engineered nano-vesicles for IR820 delivery: a therapy platform for cancer by surgery and photothermal therapy. NANOSCALE 2022; 14:2780-2792. [PMID: 35119448 DOI: 10.1039/d1nr05601h] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Long-term unsolved health problems from pre-/intra-/postoperative complications and thermal ablation complications pose threats to liver-cancer patients. To reduce the threats, we propose a multimodal-imaging guided surgical navigation system and photothermal therapy strategy to improve specific labeling, real-time monitoring and effective treatment of hepatocellular carcinoma. Using a bioengineering approach, G-Nvs@IR820, a kind of human-cell-membrane nano-vesicle, was generated with growth arrest-specific 6 (Gas6) expressed on the membrane and with near-infrared absorbing dye (IR820) loaded into it, which is proven to be an effective nanoparticle-drug-delivery system for Axl-overexpressing hepatocellular carcinoma. G-Nvs@IR820 shows excellent features in vitro and in vivo. As Gas6 binds to Axl specifically, G-Nvs@IR820 has good targeting ability to the tumor site and also has a good ability to guide the further accurate obliteration of carcinoma from adjacent normal tissue in surgery with its highly resolved fluorescence/photoacoustic/surgical-navigation signals. Moreover, the G-Nvs@IR820 represented a new perspective for photothermal therapy. Briefly, Nvs@IR820 was synthesized at a gram scale with high affinity, specificity, and safety. It has promising potential in clinical application for IGS and PTT in Axl-overexpressing hepatoma carcinoma.
Collapse
Affiliation(s)
- Xiaojie Zhang
- Department of Hepatobiliary Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, P. R. China.
- Xiamen University Research Center of Retroperitoneal Tumor Committee of Oncology Society of Chinese Medical Association, Xiamen University, Xiamen, Fujian 361102, P. R. China
| | - Changsheng Zhou
- Department of Hepatobiliary Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, P. R. China.
- Department of Hepatobiliary Surgery, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P. R. China
- Xiamen University Research Center of Retroperitoneal Tumor Committee of Oncology Society of Chinese Medical Association, Xiamen University, Xiamen, Fujian 361102, P. R. China
| | - Fanghua Wu
- Surgery department, Affiliated Fuzhou First Hospital of Fujian Medical University, Fuzhou, Fujian 350009, P. R. China.
| | - Chang Gao
- Department of Hepatobiliary Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, P. R. China.
- Xiamen University Research Center of Retroperitoneal Tumor Committee of Oncology Society of Chinese Medical Association, Xiamen University, Xiamen, Fujian 361102, P. R. China
| | - Qianqian Liu
- Department of Hepatobiliary Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, P. R. China.
- Xiamen University Research Center of Retroperitoneal Tumor Committee of Oncology Society of Chinese Medical Association, Xiamen University, Xiamen, Fujian 361102, P. R. China
| | - Peng Lv
- Department of Hepatobiliary Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, P. R. China.
- Xiamen University Research Center of Retroperitoneal Tumor Committee of Oncology Society of Chinese Medical Association, Xiamen University, Xiamen, Fujian 361102, P. R. China
| | - Ming Li
- Department of Hepatobiliary Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, P. R. China.
- Xiamen University Research Center of Retroperitoneal Tumor Committee of Oncology Society of Chinese Medical Association, Xiamen University, Xiamen, Fujian 361102, P. R. China
| | - Liyong Huang
- Surgery department, Affiliated Fuzhou First Hospital of Fujian Medical University, Fuzhou, Fujian 350009, P. R. China.
| | - Ting Wu
- Department of Hepatobiliary Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, P. R. China.
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian 361102, P. R. China.
- Xiamen University Research Center of Retroperitoneal Tumor Committee of Oncology Society of Chinese Medical Association, Xiamen University, Xiamen, Fujian 361102, P. R. China
| | - Wengang Li
- Department of Hepatobiliary Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, P. R. China.
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian 361102, P. R. China.
- Xiamen University Research Center of Retroperitoneal Tumor Committee of Oncology Society of Chinese Medical Association, Xiamen University, Xiamen, Fujian 361102, P. R. China
| |
Collapse
|
10
|
Sabarwal A, Wedel J, Liu K, Zurakowski D, Chakraborty S, Flynn E, Briscoe DM, Balan M, Pal S. A Combination therapy using an mTOR inhibitor and Honokiol effectively induces autophagy through the modulation of AXL and Rubicon in renal cancer cells and restricts renal tumor growth following organ transplantation. Carcinogenesis 2021; 43:360-370. [PMID: 34965300 PMCID: PMC9118982 DOI: 10.1093/carcin/bgab126] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/17/2021] [Accepted: 12/28/2021] [Indexed: 12/31/2022] Open
Abstract
Development of cancer, including renal cancer, is a major problem in immunosuppressed patients. The mTOR inhibitor Rapamycin (RAPA) is used as an immunosuppressive agent in patients with organ transplants and other immunological disorders; and it also has antitumorigenic potential. However, long-term use of RAPA causes reactivation of Akt, and ultimately leads to enhanced tumor growth. Honokiol (HNK) is a natural compound, which possesses both anti-inflammatory and antitumorigenic properties. In this study, we investigated the effect of a novel combination therapy using RAPA + HNK on allograft survival and post-transplantation renal tumor growth. We observed that it effectively modulated the expression of some key regulatory molecules (like Carabin, an endogenous Ras inhibitor; and Rubicon, a negative regulator of autophagy) that play important roles in tumor cell growth and survival. This combination induced toxic autophagy and apoptosis to promote cancer cell death; and was associated with a reduced expression of the tumor-promoting receptor tyrosine kinase AXL. Finally, we utilized a novel murine model to examine the effect of RAPA + HNK on post-transplantation renal tumor growth. The combination treatment prolonged the allograft survival and significantly inhibited post-transplantation tumor growth. It was associated with reduced tumor expression of Rubicon and the cytoprotective/antioxidant heme oxygenase-1 to overcome therapeutic resistance. It also downregulated the coinhibitory programmed death-1 ligand, which plays major role(s) in the immune escape of tumor cells. Together, this combination treatment has a great potential to restrict renal tumor growth in transplant recipients as well as other immunosuppressed patients.
Collapse
Affiliation(s)
- Akash Sabarwal
- Division of Nephrology, Boston Children’s Hospital, Boston, MA 02115, USA,Harvard Medical School, Boston, MA 02115, USA
| | - Johannes Wedel
- Division of Nephrology, Boston Children’s Hospital, Boston, MA 02115, USA,Harvard Medical School, Boston, MA 02115, USA
| | - Kaifeng Liu
- Division of Nephrology, Boston Children’s Hospital, Boston, MA 02115, USA,Harvard Medical School, Boston, MA 02115, USA
| | - David Zurakowski
- Division of Nephrology, Boston Children’s Hospital, Boston, MA 02115, USA,Harvard Medical School, Boston, MA 02115, USA
| | - Samik Chakraborty
- Division of Nephrology, Boston Children’s Hospital, Boston, MA 02115, USA,Harvard Medical School, Boston, MA 02115, USA
| | - Evelyn Flynn
- Division of Nephrology, Boston Children’s Hospital, Boston, MA 02115, USA,Harvard Medical School, Boston, MA 02115, USA
| | - David M Briscoe
- Division of Nephrology, Boston Children’s Hospital, Boston, MA 02115, USA,Harvard Medical School, Boston, MA 02115, USA,Transplant Research Program, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Murugabaskar Balan
- Division of Nephrology, Boston Children’s Hospital, Boston, MA 02115, USA,Harvard Medical School, Boston, MA 02115, USA
| | - Soumitro Pal
- Division of Nephrology, Boston Children’s Hospital, Boston, MA 02115, USA,Harvard Medical School, Boston, MA 02115, USA,To whom correspondence should be addressed. Division of Nephrology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA. Tel: +1 617 919 2989; Fax: +1 617 730 0365;
| |
Collapse
|
11
|
AXL Knock-Out in SNU475 Hepatocellular Carcinoma Cells Provides Evidence for Lethal Effect Associated with G2 Arrest and Polyploidization. Int J Mol Sci 2021; 22:ijms222413247. [PMID: 34948046 PMCID: PMC8708332 DOI: 10.3390/ijms222413247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/02/2021] [Accepted: 12/02/2021] [Indexed: 12/11/2022] Open
Abstract
AXL, a member of the TAM family, is a promising therapeutic target due to its elevated expression in advanced hepatocellular carcinoma (HCC), particularly in association with acquired drug resistance. Previously, RNA interference was used to study its role in cancer, and several phenotypic changes, including attenuated cell proliferation and decreased migration and invasion, have been reported. The mechanism of action of AXL in HCC is elusive. We first studied the AXL expression in HCC cell lines by real-time PCR and western blot and showed its stringent association with a mesenchymal phenotype. We then explored the role of AXL in mesenchymal SNU475 cells by CRISPR-Cas9 mediated gene knock-out. AXL-depleted HCC cells displayed drastic phenotypic changes, including increased DNA damage response, prolongation of doubling time, G2 arrest, and polyploidization in vitro and loss of tumorigenicity in vivo. Pharmacological inhibition of AXL by R428 recapitulated G2 arrest and polyploidy phenotype. These observations strongly suggest that acute loss of AXL in some mesenchymal HCC cells is lethal and points out that its inhibition may represent a druggable vulnerability in AXL-high HCC patients.
Collapse
|
12
|
Intrinsic and Extrinsic Control of Hepatocellular Carcinoma by TAM Receptors. Cancers (Basel) 2021; 13:cancers13215448. [PMID: 34771611 PMCID: PMC8582520 DOI: 10.3390/cancers13215448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/22/2021] [Accepted: 10/26/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Tyro3, Axl, and MerTK are receptor tyrosine kinases of the TAM family, which are activated by their ligands Gas6 and Protein S. TAM receptors have large physiological implications, including the removal of dead cells, activation of immune cells, and prevention of bleeding. In the last decade, TAM receptors have been suggested to play a relevant role in liver fibrogenesis and the development of hepatocellular carcinoma. The understanding of TAM receptor functions in tumor cells and their cellular microenvironment is of utmost importance to advances in novel therapeutic strategies that conquer chronic liver disease including hepatocellular carcinoma. Abstract Hepatocellular carcinoma (HCC) is the major subtype of liver cancer, showing high mortality of patients due to limited therapeutic options at advanced stages of disease. The receptor tyrosine kinases Tyro3, Axl and MerTK—belonging to the TAM family—exert a large impact on various aspects of cancer biology. Binding of the ligands Gas6 or Protein S activates TAM receptors causing homophilic dimerization and heterophilic interactions with other receptors to modulate effector functions. In this context, TAM receptors are major regulators of anti-inflammatory responses and vessel integrity, including platelet aggregation as well as resistance to chemotherapy. In this review, we discuss the relevance of TAM receptors in the intrinsic control of HCC progression by modulating epithelial cell plasticity and by promoting metastatic traits of neoplastic hepatocytes. Depending on different etiologies of HCC, we further describe the overt role of TAM receptors in the extrinsic control of HCC progression by focusing on immune cell infiltration and fibrogenesis. Additionally, we assess TAM receptor functions in the chemoresistance against clinically used tyrosine kinase inhibitors and immune checkpoint blockade in HCC progression. We finally address the question of whether inhibition of TAM receptors can be envisaged for novel therapeutic strategies in HCC.
Collapse
|
13
|
Perng DS, Hung CM, Lin HY, Morgan P, Hsu YC, Wu TC, Hsieh PM, Yeh JH, Hsiao P, Lee CY, Li YC, Wang YC, Chen YS, Lin CW. Role of autophagy-related protein in the prognosis of combined hepatocellular carcinoma and cholangiocarcinoma after surgical resection. BMC Cancer 2021; 21:828. [PMID: 34273969 PMCID: PMC8286562 DOI: 10.1186/s12885-021-08553-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 06/21/2021] [Indexed: 01/13/2023] Open
Abstract
Background Autophagy-related proteins may predict postresection overall survival (OS) and disease-free survival (DFS) in patients with combined hepatocellular carcinoma and cholangiocarcinoma (cHCC-CC). Methods We prospectively investigated how these proteins affect clinical prognosis in 40 patients who underwent hepatectomy for cHCC-CC from 2011 to 2019 at a Taiwanese hospital. Levels of autophagy-related proteins, namely LC3, Beclin-1, and p62, were immunohistochemically assessed in patient tumor and non-tumor tissues. Results We noted that LC3 expression was significantly correlated with mild clinicopathological characteristics, including macrovascular invasion, lymph node metastasis, American Joint Committee on Cancer and Barcelona Clinic Liver Cancer stages, recurrence, and mortality. Ten patient showed tumor recurrence, and 15 patients died. Postresection 5-year OS and DFS rates were 43.7 and 57.4%, respectively. Cox regression analysis showed that high intratumoral LC3 expression was significantly associated with improved OS [hazard ratio (HR; 95% confidence interval (CI)): (1.68–26.9), p = 0.007], but multiple tumors and microvascular invasion was significantly correlated with poor OS [HR (95% CI): 0.03 (0.01–0.34), p = 0.004, and 0.07 (0.01–0.46), p = 0.006, respectively]. Furthermore, high LC3 expression and cirrhosis had improved DFS [HR (95% CI): 51.3 (2.85–922), p = 0.008, and 17.9 (1.05–306), p = 0.046, respectively]. The 5-year OS and DFS rates were respectively 61.2 and 74.6% in high LC3 expression patients and 0 and 0% in those with low LC3 expression. Conclusion High LC3 expression in tumors is significantly associated with mild clinicopathological characteristics and favorable clinical prognosis in patients with cHCC-CC after resection. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-08553-6.
Collapse
Affiliation(s)
- Daw-Shyong Perng
- Division of Gastroenterology and Hepatology, E-Da Dachang Hospital, and School of Medicine, College of Medicine, I-Shou University, No. 1, Yida Road, Jiaosu Village, Yanchao District, Kaohsiung City, 82445, Taiwan.,Division of Gastroenterology and Hepatology, Department of Medicine, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan.,School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Chao-Ming Hung
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan.,Department of Surgery, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan.,Department of Surgery, E-Da Cancer Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Hung-Yu Lin
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan.,Department of Surgery, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan.,Department of Surgery, E-Da Cancer Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Paul Morgan
- Division of Gastroenterology and Hepatology, E-Da Dachang Hospital, and School of Medicine, College of Medicine, I-Shou University, No. 1, Yida Road, Jiaosu Village, Yanchao District, Kaohsiung City, 82445, Taiwan.,Division of Gastroenterology and Hepatology, Department of Medicine, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Yao-Chun Hsu
- Division of Gastroenterology and Hepatology, Department of Medicine, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan.,School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Tsung-Chin Wu
- Division of Gastroenterology and Hepatology, E-Da Dachang Hospital, and School of Medicine, College of Medicine, I-Shou University, No. 1, Yida Road, Jiaosu Village, Yanchao District, Kaohsiung City, 82445, Taiwan.,School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Pei-Min Hsieh
- Department of Surgery, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan.,Department of Surgery, E-Da Cancer Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Jen-Hao Yeh
- Division of Gastroenterology and Hepatology, E-Da Dachang Hospital, and School of Medicine, College of Medicine, I-Shou University, No. 1, Yida Road, Jiaosu Village, Yanchao District, Kaohsiung City, 82445, Taiwan.,Division of Gastroenterology and Hepatology, Department of Medicine, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan.,School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Pojen Hsiao
- Division of Gastroenterology and Hepatology, E-Da Dachang Hospital, and School of Medicine, College of Medicine, I-Shou University, No. 1, Yida Road, Jiaosu Village, Yanchao District, Kaohsiung City, 82445, Taiwan.,School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Chih-Yuan Lee
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Yu-Chan Li
- Division of Gastroenterology and Hepatology, E-Da Dachang Hospital, and School of Medicine, College of Medicine, I-Shou University, No. 1, Yida Road, Jiaosu Village, Yanchao District, Kaohsiung City, 82445, Taiwan.,School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Ya-Chin Wang
- Division of Gastroenterology and Hepatology, E-Da Dachang Hospital, and School of Medicine, College of Medicine, I-Shou University, No. 1, Yida Road, Jiaosu Village, Yanchao District, Kaohsiung City, 82445, Taiwan.,School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Yaw-Sen Chen
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan.,Department of Surgery, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Chih-Wen Lin
- Division of Gastroenterology and Hepatology, E-Da Dachang Hospital, and School of Medicine, College of Medicine, I-Shou University, No. 1, Yida Road, Jiaosu Village, Yanchao District, Kaohsiung City, 82445, Taiwan. .,Division of Gastroenterology and Hepatology, Department of Medicine, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan. .,School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan. .,School of Chinese Medicine, College of Chinese Medicine, and Research Center for Traditional Chinese Medicine, China Medical University, Taichung, Taiwan.
| |
Collapse
|
14
|
Autophagy, an accomplice or antagonist of drug resistance in HCC? Cell Death Dis 2021; 12:266. [PMID: 33712559 PMCID: PMC7954824 DOI: 10.1038/s41419-021-03553-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 02/13/2021] [Accepted: 02/17/2021] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC) is a highly lethal malignancy characterized by poor prognosis and a low 5-year survival rate. Drug treatment is proving to be effective in anti-HCC. However, only a small number of HCC patients exhibit sensitive responses, and drug resistance occurs frequently in advanced patients. Autophagy, an evolutionary process responsible for the degradation of cellular substances, is closely associated with the acquisition and maintenance of drug resistance for HCC. This review focuses on autophagic proteins and explores the intricate relationship between autophagy and cancer stem cells, tumor-derived exosomes, and noncoding RNA. Clinical trials involved in autophagy inhibition combined with anticancer drugs are also concerned.
Collapse
|
15
|
Guo JC, Wei QS, Dong L, Fang SS, Li F, Zhao Y. Prognostic Value of an Autophagy-Related Five-Gene Signature for Lower-Grade Glioma Patients. Front Oncol 2021; 11:644443. [PMID: 33768004 PMCID: PMC7985555 DOI: 10.3389/fonc.2021.644443] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/15/2021] [Indexed: 12/16/2022] Open
Abstract
Background: Molecular characteristics can be good indicators of tumor prognosis and have been introduced into the classification of gliomas. The prognosis of patients with newly classified lower-grade gliomas (LGGs, including grade 2 and grade 3 gliomas) is highly heterogeneous, and new molecular markers are urgently needed. Methods: Autophagy related genes (ATGs) were obtained from Human Autophagy Database (HADb). From the Cancer Genome Atlas (TCGA) and the Chinese Glioma Genome Atlas (CGGA), gene expression profiles including ATG expression information and patient clinical data were downloaded. Cox regression analysis, receiver operating characteristic (ROC) analysis, Kaplan–Meier analysis, random survival forest algorithm (RSFVH) and stratification analysis were performed. Results: Through univariate Cox regression analysis, we found a total of 127 ATGs associated with the prognosis of LGG patients from TCGA dataset and a total of 131 survival-related ATGs from CGGA dataset. Using TCGA dataset as the training group (n = 524), we constructed a five-ATG signature (including BAG1, BID, MAP1LC3C, NRG3, PTK6), which could divide LGG patients into two risk groups with significantly different overall survival (Log Rank P < 0.001). Then we confirmed in the independent CGGA dataset that the five-ATG signature had the ability to predict prognosis (n = 431, Log Rank P < 0.001). We further discovered that the predictive ability of the five-ATG signature was better than the existing clinical indicators and IDH mutation status. In addition, the five-ATG signature could further classify patients after receiving radiotherapy or chemotherapy into groups with different prognosis. Conclusions: We identified a five-ATG signature that could be a reliable prognostic marker and might be therapeutic targets for autophagy therapy for LGG patients.
Collapse
Affiliation(s)
- Jin-Cheng Guo
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qing-Shuang Wei
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Lei Dong
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Shuang-Sang Fang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Feng Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yi Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
16
|
Méndez-Blanco C, Fernández-Palanca P, Fondevila F, González-Gallego J, Mauriz JL. Prognostic and clinicopathological significance of hypoxia-inducible factors 1α and 2α in hepatocellular carcinoma: a systematic review with meta-analysis. Ther Adv Med Oncol 2021; 13:1758835920987071. [PMID: 33613697 PMCID: PMC7874357 DOI: 10.1177/1758835920987071] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 12/17/2020] [Indexed: 12/19/2022] Open
Abstract
Background: Hepatocellular carcinoma (HCC) is a highly recurrent tumor after resection and has been closely related to hypoxia. Hypoxia-inducible factors 1α and 2α (HIF-1α and HIF-2α) have been shown to contribute to tumor progression and therapy resistance in HCC. We evaluated the prognostic and clinicopathological significance of HIF-1α and HIF-2α in HCC patients. Methods: We systematically searched Embase, Cochrane, PubMed, Scopus and Web of Science (WOS) from inception to 1 June 2020 for studies evaluating HIF-1α and/or HIF-2α expression in HCC. Selected articles evaluate at least one factor by immunohistochemistry (IHC) in HCC patients who underwent surgical resection, and its relationship with prognosis and/or clinicopathological features. Study protocol was registered in the International Prospective Register of Systematic Reviews (PROSPERO; CDR42020191977). We meta-analyzed the data extracted or estimated according to the Parmar method employing STATA software. We evaluated the overall effect size for the hazard ratio (HR) and odds ratio (OR) with 95% confidence interval (CI), as well as heterogeneity across studies with the I2 statistic and chi-square-based Q test. Moreover, we conducted subgroup analysis when heterogeneity was substantial. Publication bias was assessed by funnel plot asymmetry and Egger’s test. Results: HIF-1α overexpression was correlated with overall survival (OS), disease-free survival (DFS)/recurrence-free survival (RFS) and clinicopathological features including Barcelona Clinic Liver Cancer (BCLC), capsule infiltration, intrahepatic metastasis, lymph node metastasis, tumor–node–metastasis (TNM), tumor differentiation, tumor number, tumor size (3 cm), vascular invasion and vasculogenic mimicry. We also detected a possible correlation of HIF-1α with alpha-fetoprotein (AFP), cirrhosis, histological grade, tumor size (5 cm) and albumin after subgroup analysis. Initially, only DFS/RFS appeared to be associated with HIF-2α overexpression. Subgroup analysis denoted that HIF-2α overexpression was related to OS and capsule infiltration. Conclusions: HIF-1α and HIF-2α overexpression is related to poor OS, DFS/RFS and some clinicopathological features of HCC patients, suggesting that both factors could be useful HCC biomarkers.
Collapse
Affiliation(s)
| | | | - Flavia Fondevila
- Institute of Biomedicine (IBIOMED), University of León, León, Spain
| | | | - José L Mauriz
- Institute of Biomedicine (IBIOMED), University of León, Campus of Vegazana s/n, León 24071, Spain
| |
Collapse
|
17
|
Bortolami M, Comparato A, Benna C, Errico A, Maretto I, Pucciarelli S, Cillo U, Farinati F. Gene and protein expression of mTOR and LC3 in hepatocellular carcinoma, colorectal liver metastasis and "normal" liver tissues. PLoS One 2020; 15:e0244356. [PMID: 33362215 PMCID: PMC7757890 DOI: 10.1371/journal.pone.0244356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 12/08/2020] [Indexed: 02/07/2023] Open
Abstract
The physiological role of autophagy in the progression of liver diseases is still debated. To understand the clinical relevance of autophagy in primary e secondary hepatic tumors, we analyzed the expression of mTOR (mammalian target of rapamycin), a key regulator of autophagy; Raptor (regulatory-associated protein of mTOR); ULK1 (Unc-51 like kinase 1) determinant in the autophagy initiation; LC3 (microtubule-associated protein 1A/1B-light chain 3), a specific marker of autophagosomes; and p62, a selective autophagy receptor. Samples from subjects with chronic hepatitis (n.58), cirrhosis (n.12), hepatocellular carcinoma (HCC, n.56), metastases (n.48) from colorectal cancer and hyperplasia or gallbladder stones (n.7), the latter considered as controls, were examined. Gene expression analysis was carried out in n.213 tissues by absolute q-PCR, while protein expression by Western Blot in n.191 lysates, including tumoral, surrounding tumoral and normal tissues. Nonparametric statistical tests were used for comparing expression levels in the above-mentioned groups. Subgroup analysis was performed considering viral infection and chemotherapy treatment. The mTOR transcriptional level was significantly lower in metastases compared to HCC (P = 0.0001). p-mTOR(Ser2448) and LC3II/LC3I protein levels were significantly higher in metastases compared to HCC (P = 0.008 and P<0.0001, respectively). ULK(Ser757) levels were significantly higher in HCC compared to metastases (P = 0.0002) while the HCV- and HBV- related HCC showed the highest p62 levels. Chemotherapy induced a down-regulation of the p-mTOR(Ser2448) in metastases and in non-tumor surrounding tissues in treated patients compared to untreated (P = 0.001 and P = 0.005, respectively). Conclusions: the different expression of proteins considered, owning their interaction and diverse tissue microenvironment, indicate an impairment of the autophagy flux in primary liver tumors that is critical for the promotion of tumorigenesis process and a coexistence of autophagy inhibition and activation mechanisms in secondary liver tumors. Differences in mTOR and LC3 transcripts emerged in tumor-free tissues, therefore particular attention should be considered in selecting the control group.
Collapse
Affiliation(s)
- Marina Bortolami
- Department of Surgery, Oncology and Gastroenterology, DISCOG, School of Medicine, Gastroenterology Unit, University of Padova, Padova, Italy
| | - Alessandra Comparato
- Department of Surgery, Oncology and Gastroenterology, DISCOG, School of Medicine, Gastroenterology Unit, University of Padova, Padova, Italy
| | - Clara Benna
- Department of Surgery, Oncology and Gastroenterology, DISCOG, School of Medicine, Surgical Unit, University of Padova, Padova, Italy
| | - Andrea Errico
- Department of Surgery, Oncology and Gastroenterology, DISCOG, School of Medicine, Gastroenterology Unit, University of Padova, Padova, Italy
| | - Isacco Maretto
- Department of Surgery, Oncology and Gastroenterology, DISCOG, School of Medicine, Surgical Unit, University of Padova, Padova, Italy
| | - Salvatore Pucciarelli
- Department of Surgery, Oncology and Gastroenterology, DISCOG, School of Medicine, Surgical Unit, University of Padova, Padova, Italy
| | - Umberto Cillo
- Department of Surgery, Oncology and Gastroenterology, DISCOG, School of Medicine, Surgical Unit, University of Padova, Padova, Italy
| | - Fabio Farinati
- Department of Surgery, Oncology and Gastroenterology, DISCOG, School of Medicine, Gastroenterology Unit, University of Padova, Padova, Italy
| |
Collapse
|
18
|
Hao TT, Liao R, Lei DL, Hu GL, Luo F. Inhibition of B7-H4 promotes hepatocellular carcinoma cell apoptosis and autophagy through the PI3K signaling pathway. Int Immunopharmacol 2020; 88:106889. [PMID: 32805693 DOI: 10.1016/j.intimp.2020.106889] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 08/06/2020] [Accepted: 08/07/2020] [Indexed: 02/07/2023]
Abstract
B7-H4 and autophagy can regulate or be induced by the PI3K signaling pathway. However, the association between B7-H4 and autophagy in hepatocellular carcinoma (HCC)remains unclear. The aim of this work was to investigate whether B7-H4 regulates autophagy via the PI3K signaling pathway in HCC cells. Here, western blotting was used to measure the expression of the related proteins involved in changes in of autophagy and apoptosis, such as LC3, P62, cleaved caspase 3, cleaved PARP, BCL-2, and BAX in Huh7 and Hep3B cells. Additionally, PI3K/AKT/mTOR signaling pathway proteins were measured. Cell counting kit-8 and flow cytometry were used to analyze the effects of B7-H4 siRNA interference on cell proliferation with the interference of B7-H4 siRNA. We found that B7-H4 siRNA increased HCC cell apoptosis and autophagy, and reduced cell proliferation. Moreover, the apoptosis-related proteins cleaved caspase 3, cleaved PARP and BAX were increased and Bcl-2 was decreased after B7-H4 siRNA interference. The expression level of the autophagy-related protein LC3Ⅱ was upregulated, while expression of the autophagy adaptor P62 expression was decreased in B7-H4 siRNA-pretreated cells. Furthermore, our data revealed that B7-H4 regulated apoptosis and autophagy through the PI3K signaling pathway in HCC cells. Therefore, these results suggested that B7-H4 plays an important role in HCC progression by affecting cell apoptosis and autophagy.
Collapse
Affiliation(s)
- Tuan-Tuan Hao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Rui Liao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Deng-Liang Lei
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Gang-Li Hu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Fang Luo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
19
|
Hsu CC, Hsieh PM, Chen YS, Lo GH, Lin HY, Dai CY, Huang JF, Chuang WL, Chen YL, Yu ML, Lin CW. Axl and autophagy LC3 expression in tumors is strongly associated with clinical prognosis of hepatocellular carcinoma patients after curative resection. Cancer Med 2019; 8:3453-3463. [PMID: 31094090 PMCID: PMC6601576 DOI: 10.1002/cam4.2229] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/10/2019] [Accepted: 04/18/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The role of Axl and LC3 as predictors of tumor recurrence and overall survival (OS) after hepatocellular carcinoma (HCC) resection remains unclear. METHODS We retrospectively included 535 HCC patients who underwent hepatectomy from 2010 to 2014 in this study. Axl and the autophagy-related marker LC3 were immunohistochemically assessed in tumors. RESULTS Axl expression was significantly associated with advanced clinicopathological features, including cirrhosis, microvascular invasion, macrovascular invasion, tumor size, BCLC stage, recurrence, and mortality. HCC recurrence occurred in 245 patients, and 219 patients died. The 5-year cumulative incidences of HCC recurrence and OS rate after HCC resection were 53.3% and 58.8%, respectively. In the Cox proportional analyses, high Axl expression and high LC3 expression were significantly associated with HCC recurrence (hazard ratio [HR]: 3.85, 95% confidence interval [CI]: 2.95-5.02, P < 0.001; and HR: 0.38, 95% CI: 0.26-0.55, P < 0.001, respectively). In addition, HCC recurrence (HR: 2.87, 95% CI: 2.01-4.01, P < 0.0001), microvascular invasion (HR: 1.85, 95% CI: 1.08-3.19, P = 0.026), hepatitis B virus-related HCC (HR: 1.77, 95% CI: 1. 21-2.56, P = 0.003), high Axl expression (HR: 1.66, 95% CI: 1.41-1.97, P < 0.0001), antiviral therapy (HR: 0.54, CI: 0.38-0.76, P < 0.001) and LC3 expression (HR: 0.41, 95% CI: 0.28-0.58, P < 0.001) were significantly associated with mortality. Furthermore, patients with a combination of high Axl and low LC3 expression had the highest risk of HCC recurrence (HR: 6.53, 95% CI: 4.11-10.4, P < 0.001) and mortality (HR: 6.66, 95% CI: 4.07-10.9, P < 0.001). In patients with high Axl, low LC3, and combined high Axl and low LC3 expression, the 5-year cumulative incidences of HCC recurrence and OS rate were 77.9%, 73.3%, and 90.0% and 28.8%, 26.7%, and 16.8%, respectively. CONCLUSION High Axl expression in tumors is associated with aggressive tumor behavior and worse clinical outcomes. Furthermore, the combination of high Axl and low LC3 expression significantly predicts poorer prognosis for HCC patients who underwent hepatectomy.
Collapse
Affiliation(s)
- Chia-Chang Hsu
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan.,Health Examination Center, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan.,Division of Gastroenterology and Hepatology, E-Da Dachang Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Pei-Min Hsieh
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan.,Department of Surgery, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Yaw-Sen Chen
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan.,Department of Surgery, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Gin-Ho Lo
- Division of Gastroenterology and Hepatology, E-Da Dachang Hospital, I-Shou University, Kaohsiung, Taiwan.,Division of Gastroenterology and Hepatology, Department of Medicine, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Hung-Yu Lin
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan.,Department of Surgery, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan.,Center for Infectious Disease and Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chia-Yen Dai
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Hepatobiliary Division, Department of Internal Medicine, and Hepatitis Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jee-Fu Huang
- Hepatobiliary Division, Department of Internal Medicine, and Hepatitis Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wan-Long Chuang
- Hepatobiliary Division, Department of Internal Medicine, and Hepatitis Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yao-Li Chen
- Division of General Surgery, Department of Surgery, Changhua Christian Hospital, Changhua, Taiwan
| | - Ming-Lung Yu
- Center for Infectious Disease and Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Hepatobiliary Division, Department of Internal Medicine, and Hepatitis Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan.,Center for Intelligent Drug Systems and Smart Bio-devices, College of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan
| | - Chih-Wen Lin
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan.,Health Examination Center, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan.,Division of Gastroenterology and Hepatology, E-Da Dachang Hospital, I-Shou University, Kaohsiung, Taiwan.,Division of Gastroenterology and Hepatology, Department of Medicine, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan.,Research Center for Traditional Chinese Medicine, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|