1
|
Wu J, Zhao X, Ren B, Duan X, Sun J. Radioresistance in Hepatocellular Carcinoma: Biological Bases and Therapeutic Implications. Int J Mol Sci 2025; 26:1839. [PMID: 40076465 PMCID: PMC11899467 DOI: 10.3390/ijms26051839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 02/13/2025] [Accepted: 02/17/2025] [Indexed: 03/14/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a malignant tumor with high morbidity and mortality. Radiotherapy technology is a common treatment modality that can be used in all stages of HCC. However, in some cases, radiotherapy fails in clinical practice mainly because of the patient's resistance to radiotherapy, creating a bottleneck for future breakthroughs. HCC radiosensitivity is primarily related to DNA double-strand break repair, cellular autophagy, cell cycle, cellular metabolism, and hypoxic environmental regulators. Therefore, a comprehensive understanding of its molecular mechanisms will be of immense importance in reversing HCC radioresistance. In this review, we provide a comprehensive overview of the mechanism of action of radiotherapy on HCC, the cellular and molecular basis of radiation resistance in HCC, related treatment modalities, and future prospects.
Collapse
Affiliation(s)
- Jianhui Wu
- Department of Radiation Oncology, The Fifth Medical Center of PLA General Hospital, Beijing 100039, China
- Medical School, Chinese PLA General Hospital, Beijing 100853, China
| | - Xiaofang Zhao
- Department of Radiation Oncology, The Fifth Medical Center of PLA General Hospital, Beijing 100039, China
- Medical School, Chinese PLA General Hospital, Beijing 100853, China
| | - Bowen Ren
- Medical School, Chinese PLA General Hospital, Beijing 100853, China
| | - Xuezhang Duan
- Department of Radiation Oncology, The Fifth Medical Center of PLA General Hospital, Beijing 100039, China
| | - Jing Sun
- Department of Radiation Oncology, The Fifth Medical Center of PLA General Hospital, Beijing 100039, China
- Medical School, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
2
|
Singh V, Pathak S, Kumar N, Jayandharan GR. Development of an Optimized Promoter System for Exosomal and Naked AAV Vector-Based Suicide Gene Therapy in Hepatocellular Carcinoma. ACS OMEGA 2024; 9:30945-30953. [PMID: 39035883 PMCID: PMC11256310 DOI: 10.1021/acsomega.4c03949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/10/2024] [Accepted: 06/14/2024] [Indexed: 07/23/2024]
Abstract
Suicide gene therapy is a promising strategy for the potential treatment of hepatocellular carcinoma (HCC). However, the lack of high transduction efficiency and targeted vectors in delivering the suicide genes to only the HCC cells is a major impediment. In the present study, we utilized an adeno-associated virus serotype 6 (AAV6) and its exosomal counterpart (exo-AAV) comprising of an inducible Caspase 9 (iCasp9) gene under the control of different promoter systems for targeting HCC cells. We employed a ubiquitous cytomegalovirus immediate early enhancer/chicken β actin promoter (CAG), a liver-specific promoter (LP1), and a baculoviral IAP repeat-containing protein 5 (BIRC5) promoter for liver and cancer cell-specific expression of iCasp9, respectively. We further evaluated these vectors in Huh7 cells for their ability to kill the target cells. BIRC5 and LP1 promoter-driven iCasp9 vectors demonstrated superior cytotoxicity when compared to CAG promoter-driven iCasp9 vectors. Further validation in a murine model of HCC demonstrated that the LP1-iCasp9 or Birc5-iCasp9-based AAV6 vectors contributed to tumor regression (∼2 fold) as effectively as the AAV6-CAG-iCasp9 vectors (∼1.9 fold). Similarly, exo-AAV6 vectors showed ∼2.1 to 2.8 fold superior in vivo tumor regression when compared to mock-treated animals. Our study has developed two novel promoters (LP1 or BIRC5) whose efficacy is comparable to a strong ubiquitous promoter in both AAV and exo-AAV systems. This expands the toolkit of AAV vectors for safe and effective treatment of HCC.
Collapse
Affiliation(s)
- Vijayata Singh
- Laurus Center for Gene Therapy,
Department of Biological Sciences and Bioengineering and Mehta Family
Center for Engineering in Medicine and Gangwal School of Medical Sciences
and Technology, Indian Institute of Technology, Kanpur, Uttar Pradesh 208016, India
| | - Subhajit Pathak
- Laurus Center for Gene Therapy,
Department of Biological Sciences and Bioengineering and Mehta Family
Center for Engineering in Medicine and Gangwal School of Medical Sciences
and Technology, Indian Institute of Technology, Kanpur, Uttar Pradesh 208016, India
| | - Narendra Kumar
- Laurus Center for Gene Therapy,
Department of Biological Sciences and Bioengineering and Mehta Family
Center for Engineering in Medicine and Gangwal School of Medical Sciences
and Technology, Indian Institute of Technology, Kanpur, Uttar Pradesh 208016, India
| | - Giridhara R. Jayandharan
- Laurus Center for Gene Therapy,
Department of Biological Sciences and Bioengineering and Mehta Family
Center for Engineering in Medicine and Gangwal School of Medical Sciences
and Technology, Indian Institute of Technology, Kanpur, Uttar Pradesh 208016, India
| |
Collapse
|
3
|
Gu X, Mu C, Zheng R, Zhang Z, Zhang Q, Liang T. The Cancer Antioxidant Regulation System in Therapeutic Resistance. Antioxidants (Basel) 2024; 13:778. [PMID: 39061847 PMCID: PMC11274344 DOI: 10.3390/antiox13070778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/15/2024] [Accepted: 06/22/2024] [Indexed: 07/28/2024] Open
Abstract
Antioxidants play a pivotal role in neutralizing reactive oxygen species (ROS), which are known to induce oxidative stress. In the context of cancer development, cancer cells adeptly maintain elevated levels of both ROS and antioxidants through a process termed "redox reprogramming". This balance optimizes the proliferative influence of ROS while simultaneously reducing the potential for ROS to cause damage to the cell. In some cases, the adapted antioxidant machinery can hamper the efficacy of treatments for neoplastic diseases, representing a significant facet of the resistance mechanisms observed in cancer therapy. In this review, we outline the contribution of antioxidant systems to therapeutic resistance. We detail the fundamental constituents of these systems, encompassing the central regulatory mechanisms involving transcription factors (of particular importance is the KEAP1/NRF2 signaling axis), the molecular effectors of antioxidants, and the auxiliary systems responsible for NADPH generation. Furthermore, we present recent clinical trials based on targeted antioxidant systems for the treatment of cancer, assessing the potential as well as challenges of this strategy in cancer therapy. Additionally, we summarize the pressing issues in the field, with the aim of illuminating a path toward the emergence of novel anticancer therapeutic approaches by orchestrating redox signaling.
Collapse
Affiliation(s)
- Xuanhao Gu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (X.G.); (C.M.); (Z.Z.)
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China;
| | - Chunyang Mu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (X.G.); (C.M.); (Z.Z.)
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China;
| | - Rujia Zheng
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China;
| | - Zhe Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (X.G.); (C.M.); (Z.Z.)
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China;
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou 310003, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou 310003, China
- Zhejiang University Cancer Center, Hangzhou 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Qi Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (X.G.); (C.M.); (Z.Z.)
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China;
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou 310003, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou 310003, China
- Zhejiang University Cancer Center, Hangzhou 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (X.G.); (C.M.); (Z.Z.)
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China;
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou 310003, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou 310003, China
- Zhejiang University Cancer Center, Hangzhou 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| |
Collapse
|
4
|
Pei R, Zhao L, Ding Y, Su Z, Li D, Zhu S, Xu L, Zhao W, Zhou W. JMJD6-BRD4 complex stimulates lncRNA HOTAIR transcription by binding to the promoter region of HOTAIR and induces radioresistance in liver cancer stem cells. J Transl Med 2023; 21:752. [PMID: 37880710 PMCID: PMC10599021 DOI: 10.1186/s12967-023-04394-y] [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: 07/24/2022] [Accepted: 10/21/2022] [Indexed: 10/27/2023] Open
Abstract
BACKGROUND Long non-coding RNA (lncRNA) HOTAIR acts importantly in liver cancer development, but its effect on radioresistance remains poorly understood. Here, our study probed into the possible impact of HOTAIR in radioresistance in liver cancer stem cells (LCSCs) and to elucidate its molecular basis. METHODS Following sorting of stem and non-stem liver cancer cells, LCSCs were identified and subjected to RNA-seq analysis for selecting differentially expressed genes. Expression of HOTAIR was determined in liver cancer tissues and CSCs. The stemness, proliferation, apoptosis and radioresistance of LCSCs were then detected in response to altered expression of HOTAIR-LSD1-JMJD6-BRD4. RESULTS Ectopic HOTAIR expression was found to promote radioresistance of LCSCs by maintaining its stemness. Mechanistic investigations indicated that HOTAIR recruited LSD1 to the MAPK1 promoter region and reduced the level of H3K9me2 in the promoter region, thus elevating ERK2 (MAPK1) expression. JMJD6-BRD4 complex promoted HOTAIR transcription by forming a complex and positively regulated ERK2 (MAPK1) expression, maintaining the stemness of LCSCs, and ultimately promoting their radioresistance in vitro and in vivo. CONCLUSION Collectively, our work highlights the promoting effect of the JMJD6-BRD4 complex on the radioresistance of LCSCs through a HOTAIR-dependent mechanism.
Collapse
Affiliation(s)
- Ruifeng Pei
- Department of Hepatopancreatobiliary Surgery, Xuzhou Cancer Hospital, Gulou District, No. 131, Huancheng Road, Xuzhou, 221005, Jiangsu, People's Republic of China
| | - Le Zhao
- Department of Hepatopancreatobiliary Surgery, Xuzhou Cancer Hospital, Gulou District, No. 131, Huancheng Road, Xuzhou, 221005, Jiangsu, People's Republic of China
| | - Yiren Ding
- Department of Hepatopancreatobiliary Surgery, Xuzhou Cancer Hospital, Gulou District, No. 131, Huancheng Road, Xuzhou, 221005, Jiangsu, People's Republic of China
| | - Zhan Su
- Department of Hepatopancreatobiliary Surgery, Xuzhou Cancer Hospital, Gulou District, No. 131, Huancheng Road, Xuzhou, 221005, Jiangsu, People's Republic of China
| | - Deqiang Li
- Department of Hepatopancreatobiliary Surgery, Xuzhou Cancer Hospital, Gulou District, No. 131, Huancheng Road, Xuzhou, 221005, Jiangsu, People's Republic of China
| | - Shuo Zhu
- Department of Hepatopancreatobiliary Surgery, Xuzhou Cancer Hospital, Gulou District, No. 131, Huancheng Road, Xuzhou, 221005, Jiangsu, People's Republic of China
| | - Lu Xu
- Department of Hepatopancreatobiliary Surgery, Xuzhou Cancer Hospital, Gulou District, No. 131, Huancheng Road, Xuzhou, 221005, Jiangsu, People's Republic of China
| | - Wei Zhao
- Department of Hepatopancreatobiliary Surgery, Xuzhou Cancer Hospital, Gulou District, No. 131, Huancheng Road, Xuzhou, 221005, Jiangsu, People's Republic of China.
| | - Wuyuan Zhou
- Department of Hepatopancreatobiliary Surgery, Xuzhou Cancer Hospital, Gulou District, No. 131, Huancheng Road, Xuzhou, 221005, Jiangsu, People's Republic of China.
| |
Collapse
|
5
|
Yang Y, Xiong L, Li M, Jiang P, Wang J, Li C. Advances in radiotherapy and immunity in hepatocellular carcinoma. J Transl Med 2023; 21:526. [PMID: 37542324 PMCID: PMC10401766 DOI: 10.1186/s12967-023-04386-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/24/2023] [Indexed: 08/06/2023] Open
Abstract
Primary liver cancer is one of the most common malignant tumours worldwide; it caused approximately 830,000 deaths in 2020. Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer, accounting for over 80% of all cases. Various methods, including surgery, chemotherapy, radiotherapy, and radiofrequency ablation, have been widely used in the treatment of HCC. With the advancement of technology, radiotherapy has become increasingly important in the comprehensive treatment of HCC. However, due to the insufficient sensitivity of tumour cells to radiation, there are still multiple limitation in clinical application of radiotherapy. In recent years, the role of immunotherapy in cancer has been increasingly revealed, and more researchers have turned their attention to the combined application of immunotherapy and radiotherapy in the hope of achieving better treatment outcomes. This article reviews the progress on radiation therapy in HCC and the current status of its combined application with immunotherapy, and discusses the prospects and value of radioimmunotherapy in HCC.
Collapse
Affiliation(s)
- Yuhan Yang
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, 100191, China
| | - Liting Xiong
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, 100191, China
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China
| | - Mengyuan Li
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, 100191, China
| | - Ping Jiang
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, 100191, China.
| | - Junjie Wang
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, 100191, China.
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China.
| | - Chunxiao Li
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
6
|
Gupta R, Kadhim MM, Turki Jalil A, Obayes AM, Aminov Z, Alsaikhan F, Ramírez-Coronel AA, Ramaiah P, Tayyib NA, Luo X. Multifaceted role of NF-κB in hepatocellular carcinoma therapy: Molecular landscape, therapeutic compounds and nanomaterial approaches. ENVIRONMENTAL RESEARCH 2023; 228:115767. [PMID: 36966991 DOI: 10.1016/j.envres.2023.115767] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 05/16/2023]
Abstract
The predominant kind of liver cancer is hepatocellular carcinoma (HCC) that its treatment have been troublesome difficulties for physicians due to aggressive behavior of tumor cells in proliferation and metastasis. Moreover, stemness of HCC cells can result in tumor recurrence and angiogenesis occurs. Another problem is development of resistance to chemotherapy and radiotherapy in HCC cells. Genomic mutations participate in malignant behavior of HCC and nuclear factor-kappaB (NF-κB) has been one of the oncogenic factors in different human cancers that after nuclear translocation, it binds to promoter of genes in regulating their expression. Overexpression of NF-κB has been well-documented in increasing proliferation and invasion of tumor cells and notably, when its expression enhances, it induces chemoresistance and radio-resistance. Highlighting function of NF-κB in HCC can shed some light on the pathways regulating progression of tumor cells. The first aspect is proliferation acceleration and apoptosis inhibition in HCC cells mediated by enhancement in expression level of NF-κB. Moreover, NF-κB is able to enhance invasion of HCC cells via upregulation of MMPs and EMT, and it triggers angiogenesis as another step for increasing spread of tumor cells in tissues and organs. When NF-κB expression enhances, it stimulates chemoresistance and radio-resistance in HCC cells and by increasing stemness and population of cancer-stem cells, it can provide the way for recurrence of tumor. Overexpression of NF-κB mediates therapy resistance in HCC cells and it can be regulated by non-coding RNAs in HCC. Moreover, inhibition of NF-κB by anti-cancer and epigenetic drugs suppresses HCC tumorigenesis. More importantly, nanoparticles are considered for suppressing NF-κB axis in cancer and their prospectives and results can also be utilized for treatment of HCC. Nanomaterials are promising factors in treatment of HCC and by delivery of genes and drugs, they suppress HCC progression. Furthermore, nanomaterials provide phototherapy in HCC ablation.
Collapse
Affiliation(s)
- Reena Gupta
- Institute of Pharmaceutical Research, GLA University, District-Mathura, U. P., India
| | - Mustafa M Kadhim
- Department of Dentistry, Kut University College, Kut, Wasit, 52001, Iraq; Medical Laboratory Techniques Department, Al-Farahidi University, Baghdad, 10022, Iraq
| | - Abduladheem Turki Jalil
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq.
| | | | - Zafar Aminov
- Department of Public Health and Healthcare Management, Samarkand State Medical University, 18 Amir Temur Street, Samarkand, Uzbekistan; Department of Scientific Affairs, Tashkent State Dental Institute, 103 Makhtumkuli Str., Tashkent, Uzbekistan
| | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia.
| | - Andrés Alexis Ramírez-Coronel
- Azogues Campus Nursing Career, Health and Behavior Research Group (HBR), Psychometry and Ethology Laboratory, Catholic University of Cuenca, Ecuador; Epidemiology and Biostatistics Research Group, CES University, Colombia; Educational Statistics Research Group (GIEE), National University of Education, Ecuador
| | | | - Nahla A Tayyib
- Faculty of Nursing, Umm al- Qura University, Makkah, Saudi Arabia
| | - Xuanming Luo
- Department of General Surgery, Shanghai Xuhui Central Hospital, Fudan University, Shanghai, 200031, China.
| |
Collapse
|
7
|
Hashemi M, Nadafzadeh N, Imani MH, Rajabi R, Ziaolhagh S, Bayanzadeh SD, Norouzi R, Rafiei R, Koohpar ZK, Raei B, Zandieh MA, Salimimoghadam S, Entezari M, Taheriazam A, Alexiou A, Papadakis M, Tan SC. Targeting and regulation of autophagy in hepatocellular carcinoma: revisiting the molecular interactions and mechanisms for new therapy approaches. Cell Commun Signal 2023; 21:32. [PMID: 36759819 PMCID: PMC9912665 DOI: 10.1186/s12964-023-01053-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 01/15/2023] [Indexed: 02/11/2023] Open
Abstract
Autophagy is an evolutionarily conserved process that plays a role in regulating homeostasis under physiological conditions. However, dysregulation of autophagy is observed in the development of human diseases, especially cancer. Autophagy has reciprocal functions in cancer and may be responsible for either survival or death. Hepatocellular carcinoma (HCC) is one of the most lethal and common malignancies of the liver, and smoking, infection, and alcohol consumption can lead to its development. Genetic mutations and alterations in molecular processes can exacerbate the progression of HCC. The function of autophagy in HCC is controversial and may be both tumor suppressive and tumor promoting. Activation of autophagy may affect apoptosis in HCC and is a regulator of proliferation and glucose metabolism. Induction of autophagy may promote tumor metastasis via induction of EMT. In addition, autophagy is a regulator of stem cell formation in HCC, and pro-survival autophagy leads to cancer cell resistance to chemotherapy and radiotherapy. Targeting autophagy impairs growth and metastasis in HCC and improves tumor cell response to therapy. Of note, a large number of signaling pathways such as STAT3, Wnt, miRNAs, lncRNAs, and circRNAs regulate autophagy in HCC. Moreover, regulation of autophagy (induction or inhibition) by antitumor agents could be suggested for effective treatment of HCC. In this paper, we comprehensively review the role and mechanisms of autophagy in HCC and discuss the potential benefit of targeting this process in the treatment of the cancer. Video Abstract.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Niloufar Nadafzadeh
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad Hassan Imani
- Department of Clinical Science, Faculty of Veterinary Medicine, Shahr-E Kord Branch, Islamic Azad University, Tehran, Chaharmahal and Bakhtiari, Iran
| | - Romina Rajabi
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Setayesh Ziaolhagh
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | | | - Raheleh Norouzi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Reihaneh Rafiei
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Zeinab Khazaei Koohpar
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| | - Behnaz Raei
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohammad Arad Zandieh
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
- Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, Australia
- AFNP Med Austria, Vienna, Austria
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Shing Cheng Tan
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
8
|
Ligaza γ-glutamylocysteiny – od molekularnych mechanizmów regulacji aktywności enzymatycznej do implikacji terapeutycznych. POSTEP HIG MED DOSW 2021. [DOI: 10.2478/ahem-2021-0013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Abstrakt
Glutation (γ-glutamylocysteinyloglicyna, GSH) jest najbardziej rozpowszechnionym tiolowym antyoksydantem wytwarzanym w cytozolu wszystkich komórek ssaków, który pełni ważną rolę ochronną przed stresem oksydacyjnym. GSH jest syntetyzowany de novo przez sekwencyjne działanie dwóch enzymów: ligazy γ-glutamylocysteiny (GCL) i syntetazy glutationowej (GS). GCL katalizuje pierwszy etap biosyntezy GSH, którego produktem jest γ-glutamylocysteina (γ-GC). GCL jest heterodimerycznym enzymem zbudowanym z podjednostki katalitycznej (GCLc) i modulatorowej (GCLm), kodowanych przez dwa różne geny. Podjednostki GCL podlegają złożonej regulacji zarówno na poziomie przed-, jak i potranslacyjnym. Zmiany w ekspresji i aktywności GCL mogą zaburzać poziom GSH i homeostazy redoks. Przyczyną wielu przewlekłych schorzeń związanych ze stresem oksydacyjnym jest upośledzenie aktywności katalitycznej GCL oraz spadek stężenia GSH. Badania przedkliniczne sugerują, że podawanie egzogennej γ-GC podwyższa wewnątrzkomórkowe GSH przez dostarczenie brakującego substratu i może wykazywać potencjał jako terapia uzupełniająca w chorobach związanych z deplecją GSH.
Collapse
|
9
|
Zhu Y, Sui B, Liu X, Sun J. The reversal of drug resistance by two-dimensional titanium carbide Ti 2 C (2D Ti2C) in non-small-cell lung cancer via the depletion of intracellular antioxidant reserves. Thorac Cancer 2021; 12:3340-3355. [PMID: 34741403 PMCID: PMC8671908 DOI: 10.1111/1759-7714.14208] [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: 07/12/2021] [Revised: 10/09/2021] [Accepted: 10/11/2021] [Indexed: 12/22/2022] Open
Abstract
Background Chemoresistance is a major barrier limiting the therapeutic efficacy of late stage non‐small cell lung cancer (NSCLC). In this study, we sought to use two‐dimensional titanium carbide (2D Ti2C) to reverse cisplatin resistance in NSCLC. Methods We first achieved favorable properties as a potential anti‐tumor agent. We then compared cell viability and cisplatin uptake in chemoresistant NSCLC cells before and after the use of 2D Ti2C. Afterwards, we explored the effects of 2D Ti2C on intracellular antioxidant reserves, followed by evaluating the subsequent changes in the expression of core drug resistance genes. Finally, we confirmed the tumor inhibitory effect and bio‐safety of 2D Ti2C in a drug‐resistant lung cancer model in nude mice. Results Due to the properties of thin layer, large specific surface area, and abundant reactive groups on the surface, 2D Ti2C can deplete the antioxidant reserve systems such as the glutathione redox buffer system, γ‐glutamylcysteine synthetase (γ‐GCS), glutathione peroxidase (GPx), glutathione‐S‐transferase‐Pi (GST‐π), and metallothionein (MT), thereby increasing the intracellular accumulation of cisplatin and decreasing the expression of drug resistance genes. Conclusions 2D Ti2C can reverse NSCLC chemoresistance both in vitro and in vivo, suggesting that it may potentially become a novel and effective means to treat chemoresistant NSCLC in the clinic.
Collapse
Affiliation(s)
- Yue Zhu
- Department of Dental Materials, Shanghai Biomaterials Research & Testing Center, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center of Stomatology, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Baiyan Sui
- Department of Dental Materials, Shanghai Biomaterials Research & Testing Center, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center of Stomatology, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Xin Liu
- Department of Dental Materials, Shanghai Biomaterials Research & Testing Center, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center of Stomatology, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Jiao Sun
- Department of Dental Materials, Shanghai Biomaterials Research & Testing Center, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center of Stomatology, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| |
Collapse
|
10
|
Lin J, Ruan J, Zhu H, Chen Z, Chen J, Yu H. Tenacissoside H Induces Autophagy and Radiosensitivity of Hepatocellular Carcinoma Cells by PI3K/Akt/mTOR Signaling Pathway. Dose Response 2021; 19:15593258211011023. [PMID: 34035782 PMCID: PMC8127767 DOI: 10.1177/15593258211011023] [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: 11/09/2020] [Revised: 03/10/2021] [Accepted: 03/25/2021] [Indexed: 12/23/2022] Open
Abstract
Tenacissoside H (TEH), which has anti-inflammatory and anti-tumor effects, is a major active ingredient extracted from the stem of Marsdenia tenacissima. However, the effect of TEH on hepatocellular carcinoma (HCC) as well as the underlying mechanisms are still indistinct. Presently, HCC cells (including Huh-7 and HepG2) were dealt with different concentrations of TEH. The proliferation and apoptosis of HCC cells were determined via Cell Counting Kit-8 (CCK8) assay and flow cytometry. In addition, Western blot was conducted to evaluate the expressions of autophagy—and apoptosis-related proteins. Tissue immunofluorescence was carried out to evaluate LC3B expression in the tumor tissues. The data showed that TEH suppressed the growth of HCC cells in a concentration-dependent manner. Besides, TEH enhanced radiosensitivity and promoted the apoptosis of HCC cells. Moreover, the mRNA and protein levels of autophagy-related genes (LC3-II/LC2-I, ATG5, Beclin-1) were significantly promoted by TEH. Mechanistically, TEH attenuated the activation of PI3K/Akt/mTOR signaling pathway. However, inhibition of PI3 K pathway abolished the anti-tumor effects of TEH in HCC cells. Collectively, this study suggested that TEH increases the radiosensitivity of HCC cells via inducing autophagy and apoptosis through downregulating PI3K/Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Jiatian Lin
- Department of Minimally Invasive Intervention, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Jiyin Ruan
- Department of Minimally Invasive Intervention, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Hao Zhu
- Department of Minimally Invasive Intervention, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Zaizhong Chen
- Department of Minimally Invasive Intervention, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Junhui Chen
- Department of Minimally Invasive Intervention, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Hongjian Yu
- Department of Minimally Invasive Intervention, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
11
|
Tao H, Ge G, Liang X, Zhang W, Sun H, Li M, Geng D. ROS signaling cascades: dual regulations for osteoclast and osteoblast. Acta Biochim Biophys Sin (Shanghai) 2020; 52:1055-1062. [PMID: 33085739 DOI: 10.1093/abbs/gmaa098] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/16/2020] [Accepted: 07/30/2020] [Indexed: 02/04/2023] Open
Abstract
Accumulating evidence indicates that intracellular reactive oxygen species (ROS) production is highly involved in bone homeostasis by intervening osteoclast or osteoblast differentiation. Interestingly, ROS that are known as oxidizing agents exert dose-dependent biphasic properties in bone remodeling, including preventing osteoblast activity but accelerating osteoclast resorption. ROS mainly composed of superoxide anion radical, hydroxyl radical, nitric oxide, and two-electron reduction product hydrogen peroxide, which are important components to regulate bone cell metabolism and function in mammal skeleton. These free radicals can be partly produced in bone and boosted in an inflammation state. Although numerous researches have emphasized the impacts of ROS on bone cell biology and verified the mechanism of ROS signaling cascades, the recapitulatory commentary is necessary. In this review article, we particularly focus on the regulation of the intracellular ROS and its potential mechanism impacting on cell-signaling transduction in osteoclast and osteoblast differentiation for preferable understanding the pathogenesis and searching for novel therapeutic protocols for human bone diseases.
Collapse
Affiliation(s)
- Huaqiang Tao
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215006, China, and
| | - Gaoran Ge
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215006, China, and
| | - Xiaolong Liang
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215006, China, and
| | - Weicheng Zhang
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215006, China, and
| | - Houyi Sun
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215006, China, and
| | - Meng Li
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215006, China, and
- Division of Life Sciences and Medicine, the First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei 230000, China
| | - Dechun Geng
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215006, China, and
| |
Collapse
|
12
|
Matadamas-Guzman M, Zazueta C, Rojas E, Resendis-Antonio O. Analysis of Epithelial-Mesenchymal Transition Metabolism Identifies Possible Cancer Biomarkers Useful in Diverse Genetic Backgrounds. Front Oncol 2020; 10:1309. [PMID: 32850411 PMCID: PMC7406688 DOI: 10.3389/fonc.2020.01309] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 06/23/2020] [Indexed: 12/17/2022] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) relates to many molecular and cellular alterations that occur when epithelial cells undergo a switch in differentiation generating mesenchymal-like cells with newly acquired migratory and invasive properties. In cancer cells, EMT leads to drug resistance and metastasis. Moreover, differences in genetic backgrounds, even between patients with the same type of cancer, also determine resistance to some treatments. Metabolic rewiring is essential to induce EMT, hence it is important to identify key metabolic elements for this process, which can be later used to treat cancer cells with different genetic backgrounds. Here we used a mathematical modeling approach to determine which are the metabolic reactions altered after induction of EMT, based on metabolomic and transcriptional data of three non-small cell lung cancer (NSCLC) cell lines. The model suggested that the most affected pathways were the Krebs cycle, amino acid metabolism, and glutathione metabolism. However, glutathione metabolism had many alterations either on the metabolic reactions or at the transcriptional level in the three cell lines. We identified Glutamate-cysteine ligase (GCL), a key enzyme of glutathione synthesis, as an important common feature that is dysregulated after EMT. Analyzing survival data of men with lung cancer, we observed that patients with mutations in GCL catalytic subunit (GCLC) or Glutathione peroxidase 1 (GPX1) genes survived less time than people without mutations on these genes. Besides, patients with low expression of ANPEP, GPX3 and GLS genes also survived less time than those with high expression. Hence, we propose that glutathione metabolism and glutathione itself could be good targets to delay or potentially prevent EMT induction in NSCLC cell lines.
Collapse
Affiliation(s)
- Meztli Matadamas-Guzman
- Programa de Doctorado en Ciencias Biomédicas, UNAM, Mexico City, Mexico.,Human Systems Biology Lab, National Institute of Genomic Medicine, Mexico City, Mexico
| | - Cecilia Zazueta
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología-Ignacio Chávez, Mexico City, Mexico
| | - Emilio Rojas
- Department of Genomic Medicine and Environmental Toxicology, Institute of Biomedical Research, UNAM, Mexico City, Mexico
| | - Osbaldo Resendis-Antonio
- Human Systems Biology Lab, National Institute of Genomic Medicine, Mexico City, Mexico.,Coordinación de la Investigación Científica-Red de Apoyo a la Investigación, UNAM, Mexico City, Mexico
| |
Collapse
|
13
|
Hayes JD, Dinkova-Kostova AT, Tew KD. Oxidative Stress in Cancer. Cancer Cell 2020; 38:167-197. [PMID: 32649885 PMCID: PMC7439808 DOI: 10.1016/j.ccell.2020.06.001] [Citation(s) in RCA: 1448] [Impact Index Per Article: 289.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 04/29/2020] [Accepted: 05/29/2020] [Indexed: 12/13/2022]
Abstract
Contingent upon concentration, reactive oxygen species (ROS) influence cancer evolution in apparently contradictory ways, either initiating/stimulating tumorigenesis and supporting transformation/proliferation of cancer cells or causing cell death. To accommodate high ROS levels, tumor cells modify sulfur-based metabolism, NADPH generation, and the activity of antioxidant transcription factors. During initiation, genetic changes enable cell survival under high ROS levels by activating antioxidant transcription factors or increasing NADPH via the pentose phosphate pathway (PPP). During progression and metastasis, tumor cells adapt to oxidative stress by increasing NADPH in various ways, including activation of AMPK, the PPP, and reductive glutamine and folate metabolism.
Collapse
Affiliation(s)
- John D Hayes
- Division of Cellular Medicine, Jacqui Wood Cancer Centre, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK, Scotland.
| | - Albena T Dinkova-Kostova
- Division of Cellular Medicine, Jacqui Wood Cancer Centre, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK, Scotland; Department of Pharmacology and Molecular Sciences and Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kenneth D Tew
- Department of Cell and Molecular Pharmacology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
14
|
Kou L, Jiang X, Huang H, Lin X, Zhang Y, Yao Q, Chen R. The role of transporters in cancer redox homeostasis and cross-talk with nanomedicines. Asian J Pharm Sci 2020; 15:145-157. [PMID: 32373196 PMCID: PMC7193452 DOI: 10.1016/j.ajps.2020.02.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/03/2019] [Accepted: 02/12/2020] [Indexed: 02/07/2023] Open
Abstract
Tumor cell usually exhibits high levels of reactive oxygen species and adaptive antioxidant system due to the metabolic, genetic, and microenvironment-associated alterations. The altered redox homeostasis can promote tumor progression, development, and treatment resistance. Several membrane transporters are involved in the resetting redox homeostasis and play important roles in tumor progression. Therefore, targeting the involved transporters to disrupt the altered redox balance emerges as a viable strategy for cancer therapy. In addition, nanomedicines have drawn much attention in the past decades. Using nanomedicines to target or reset the redox homeostasis alone or combined with other therapies has brought convincing data in cancer treatment. In this review, we will introduce the altered redox balance in cancer metabolism and involved transporters, and highlight the recent advancements of redox-modulating nanomedicines for cancer treatment.
Collapse
Affiliation(s)
- Longfa Kou
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Xinyu Jiang
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Huirong Huang
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Xinlu Lin
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Youting Zhang
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Qing Yao
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- School of Pharmaceutical Sciences, Wenzhou Medical University, Chashan, Wenzhou 325035, China
- Corresponding author. Wenzhou Medical University, University Town, Wenzhou 325035, China. Tel: +86 18958969225
| | - Ruijie Chen
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Corresponding author. Wenzhou Medical University, 109 Xueyuan West Road, Wenzhou 325027, China. Tel: +86 13806890233
| |
Collapse
|
15
|
mTOR-Mediated Antioxidant Activation in Solid Tumor Radioresistance. JOURNAL OF ONCOLOGY 2019; 2019:5956867. [PMID: 31929797 PMCID: PMC6942807 DOI: 10.1155/2019/5956867] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/20/2019] [Accepted: 11/30/2019] [Indexed: 12/27/2022]
Abstract
Radiotherapy is widely used for the treatment of cancer patients, but tumor radioresistance presents serious therapy challenges. Tumor radioresistance is closely related to high levels of mTOR signaling in tumor tissues. Therefore, targeting the mTOR pathway might be a strategy to promote solid tumor sensitivity to ionizing radiation. Radioresistance is associated with enhanced antioxidant mechanisms in cancer cells. Therefore, examination of the relationship between mTOR signaling and antioxidant mechanism-linked radioresistance is required for effective radiotherapy. In particular, the effect of mTOR signaling on antioxidant glutathione induction by the Keap1-NRF2-xCT pathway is described in this review. This review is expected to assist in the identification of therapeutic adjuvants to increase the efficacy of radiotherapy.
Collapse
|
16
|
Song W, Zhang J, Xia Q, Sun M. Down-regulated lncRNA TP73-AS1 reduces radioresistance in hepatocellular carcinoma via the PTEN/Akt signaling pathway. Cell Cycle 2019; 18:3177-3188. [PMID: 31564201 DOI: 10.1080/15384101.2019.1671089] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Objective: Recently, the role of long non-coding RNAs (lncRNAs) in hepatocellular carcinoma (HCC) has been assessed. Our research was determined to investigate the impacts of lncRNA TP73-AS1 on radioresistance of HCC by modulating PTEN/Akt signaling pathway. Methods: Expression of TP73-AS1 in HCC tissues and cells was detected using reverse transcription quantitative polymerase chain reaction (RT-qPCR). The HCC cells were conducted with different doses of irradiation, then the survival, colony formation and apoptosis were determined by a series of assays. The HCC cell line with a higher expression of TP73-AS1 was transfected with TP73-AS1-siRNA and X-rayed, the expression of TP73-AS1, cell survival, radiosensitivity, and apoptosis were evaluated. Subcutaneous tumorigenesis in nude mice was adopted to record the size of tumors before and after the radiation. RT-qPCR and Western blot analysis were used to clarify the activation of PTEN/Akt signaling pathway. Results: TP73-AS1 was highly expressed in HCC tissues and cells. With the increasing dose of radiation, the relative proliferation activity and survival fraction (SF) of HCC cells was gradually reduced, while the total apoptosis rate was gradually elevated. TP73-AS1 knockdown promoted radiosensitivity and apoptosis, repressed cell proliferation, making it an inhibitor of tumor in HCC. Moreover, reduced TP73-AS1 was able to decline the phosphorylation of Akt and increase the expression of PTEN in HCC. Down-regulated TP73-AS1 could repress tumorigenesis by promoting radiosensitivity in nude mice with HCC. Conclusion: Our study suggests that lncRNA TP73-AS1 was highly expressed in HCC and participated in radioresistance of HCC via PTEN/Akt signaling pathway. Abbreviations: lncRNAs: long non-coding RNAs; lncRNAs: HCC: hepatocellular carcinoma; RT-qPCR: reverse transcription quantitative polymerase chain reaction; survival fraction: SF; lncRNA TP73-AS1: LncRNA P73 antisense RNA 1T; PTEN: Phosphatase and tensin homologue; Akt: Protein kinase B; P13K: phosphatidylinositol 3-kinase; TNM: tumor, node and metastasis; ACJJ: American Joint Committee on Cancer; FBS: fetal bovine serum; EDTA: ethylene diamine tetraacetic acid; NC: negative control; DMEM: Dulbecco's modified Eagle medium; OD: optical density; PE: Plating efficiency; FITC/PI: fluoresceine isothiocyanate/propidium iodide; PBS: phosphate buffered solution; GAPDH: Glyceraldehyde phosphate dehydrogenase; ANOVA: one-way analysis of variance; LSD-t: least significant difference test.
Collapse
Affiliation(s)
- Wei Song
- Department of Pathology, The Affiliated Cancer Hospital of Zhengzhou University , Zhengzhou , Henan , PR China
| | - Jingjing Zhang
- Department of Cardiovascularology, The Zhengzhou Central Hospital Affiliated to Zhengzhou University , Zhengzhou , Henan , PR China
| | - Qingxin Xia
- Department of Pathology, The Affiliated Cancer Hospital of Zhengzhou University , Zhengzhou , Henan , PR China
| | - Miaomiao Sun
- Department of Pathology, The Affiliated Cancer Hospital of Zhengzhou University , Zhengzhou , Henan , PR China
| |
Collapse
|
17
|
Sun J, Zhou C, Ma Q, Chen W, Atyah M, Yin Y, Fu P, Liu S, Hu B, Ren N, Zhou H. High GCLC level in tumor tissues is associated with poor prognosis of hepatocellular carcinoma after curative resection. J Cancer 2019; 10:3333-3343. [PMID: 31293636 PMCID: PMC6603424 DOI: 10.7150/jca.29769] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 05/05/2019] [Indexed: 12/21/2022] Open
Abstract
Glutamate-cysteine ligase catalytic subunit (GCLC) has been reported to overexpress in a variety types of cancer and be related with tumor progression and drug resistance. However, little has been known about GCLC's prognostic significance and biological roles in hepatocellular carcinoma (HCC). In the present study, we evaluated GCLC expression level using immunohistochemical staining (IHC) in tissue microarray (TMA) containing paired tumor and peritumoral liver tissues from 168 patients with HCC who received curative resection. GCLC levels in tumor tissues were significantly higher than in peritumoral liver tissues, and tumor GCLC level was associated with overall survival (OS) and disease-free survival (DFS). Five-year OS and DFS rates were 41.15% and 25.88% for the group with high tumor GCLC level, compared with 68.09% and 47.51% for the group with low tumor GCLC level (P<0.001 and P=0.001, respectively). Moreover, quantitative reverse transcription PCR (qRT-PCR) analysis demonstrated that GCLC was transcriptionally activated in HCC tissues when comparing with peritumoral tissues. Tumor GCLC level, which correlated to tumor differentiation, microvascular invasion and BCLC stage, was independent prognostic factors for both OS (P=0.006) and DFS (P=0.003). Importantly, tumor GCLC level was still significantly associated with OS and DFS in patients with early HCC. GCLC-based nomogram models were further established and exhibit significantly higher predictive accuracy as compared with routine clinical staging systems. In conclusion, tumor GCLC is a potential prognostic biomarker for HCC patients after receiving curative resection.
Collapse
Affiliation(s)
- Jialei Sun
- Liver Cancer Institute & Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Key Laboratory of Carcinogenesis & Cancer Invasion, Ministry of Education, China
| | - Chenhao Zhou
- Key Laboratory of Carcinogenesis & Cancer Invasion, Ministry of Education, China.,Department of Liver Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qianni Ma
- Liver Cancer Institute & Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Key Laboratory of Carcinogenesis & Cancer Invasion, Ministry of Education, China
| | - Wanyong Chen
- Department of Liver Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Department of Surgery, Institute of Fudan-Minhang Acadamic Health System, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Manar Atyah
- Department of Liver Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yirui Yin
- Department of Liver Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Peiyao Fu
- Department of Liver Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Shuang Liu
- Department of Liver Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Bo Hu
- Department of Liver Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ning Ren
- Department of Liver Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Department of Surgery, Institute of Fudan-Minhang Acadamic Health System, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Haijun Zhou
- Liver Cancer Institute & Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Key Laboratory of Carcinogenesis & Cancer Invasion, Ministry of Education, China
| |
Collapse
|
18
|
Cui Q, Wang JQ, Assaraf YG, Ren L, Gupta P, Wei L, Ashby CR, Yang DH, Chen ZS. Modulating ROS to overcome multidrug resistance in cancer. Drug Resist Updat 2018; 41:1-25. [DOI: 10.1016/j.drup.2018.11.001] [Citation(s) in RCA: 273] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 10/26/2018] [Accepted: 11/02/2018] [Indexed: 02/07/2023]
|