1
|
Wong MMT, Aziz NA, Ch'ng ES, Armon S, Chook JB, Bong JJ, Peh SC, Wu YS, Teow SY. Expression of LC3A, LC3B and p62/SQSTM1 autophagy proteins in hepatocellular carcinoma (HCC) tissues and the predicted microRNAs involved in the autophagy-related pathway. J Mol Histol 2024; 55:317-328. [PMID: 38630414 DOI: 10.1007/s10735-024-10191-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 03/31/2024] [Indexed: 05/21/2024]
Abstract
BACKGROUND Autophagy plays multifaceted roles in regulating hepatocellular carcinoma (HCC) and the mechanisms involved are under-explored. Regulatory microRNAs (miRNAs) have been reported to target autophagy proteins but their roles in HCC is not well studied. Using HCC patient tissues, this study aims to investigate the association of autophagy with several clinicopathological parameters as well as identifying the autophagy-related miRNAs and the possible pathways. METHODS AND RESULTS Autophagy level in the HCC patient-derived cancer and non-cancer tissues was determined by immunohistochemistry (IHC) targeting SQSTM1, LC3A and LC3B proteins. Significance tests of clinicopathological variables were tested using the Fisher's exact or Chi-square tests. Gene and miRNA expression assays were carried out and analyzed using Nanostring platform and software followed by validation of other online bioinformatics tools, namely String and miRabel. Autophagy expression was significantly higher in cancerous tissues compared to adjacent non-cancer tissues. High LC3B expression was associated with advanced tumor histology grade and tumor location. Nanostring gene expression analysis revealed that SQSTM1, PARP1 and ATG9A genes were upregulated in HCC tissues compared to non-cancer tissues while SIRT1 gene was downregulated. These genes are closely related to an autophagy pathway in HCC. Further, using miRabel tool, three downregulated miRNAs (hsa-miR-16b-5p, hsa-miR-34a-5p, and hsa-miR-660-5p) and one upregulated miRNA (hsa-miR-539-5p) were found to closely interact with the abovementioned autophagy-related genes. We then mapped out the possible pathway involving the genes and miRNAs in HCC tissues. CONCLUSIONS We conclude that autophagy events are more active in HCC tissues compared to the adjacent non-cancer tissues. We also reported the possible role of several miRNAs in regulating autophagy-related genes in the autophagy pathway in HCC. This may contribute to the development of potential therapeutic targets for improving HCC therapy. Future investigations are warranted to validate the target genes reported in this study using a larger sample size and more targeted molecular technique.
Collapse
Affiliation(s)
- Magdelyn Mei-Theng Wong
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Jalan University, Bandar Sunway, Selangor Darul Ehsan, Subang Jaya, 47500, Malaysia
| | - Norazlin Abdul Aziz
- Molecular Pathology Unit, Cancer Research Centre, Institute for Medical Research, National Institutes of Health (NIH Complex), Ministry of Health Malaysia, Level 4, Block C7, No: 1, Jalan Setia Murni U13/52, Section U13, Setia Alam, Selangor Darul Ehsan, Shah Alam, 40170, Malaysia
| | - Ewe Seng Ch'ng
- Advanced Medical and Dental Institute, University Sains Malaysia, 13200 Kepala Batas, Pulau Pinang, Bertam, Malaysia
| | - Subasri Armon
- Pathology Department, Hospital Kuala Lumpur, Jalan Pahang, Kuala Lumpur, 50588, Malaysia
| | - Jack-Bee Chook
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Jalan University, Bandar Sunway, Selangor Darul Ehsan, Subang Jaya, 47500, Malaysia
| | - Jan-Jin Bong
- Sunway Medical Centre, 5 Jalan Lagoon Selatan, Bandar Sunway, Subang Jaya, 47500, Selangor, Malaysia
| | - Suat-Cheng Peh
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Jalan University, Bandar Sunway, Selangor Darul Ehsan, Subang Jaya, 47500, Malaysia
- Sunway Medical Centre, 5 Jalan Lagoon Selatan, Bandar Sunway, Subang Jaya, 47500, Selangor, Malaysia
| | - Yuan Seng Wu
- Sunway Microbiome Centre, School of Medical and Life Sciences, Sunway University, Selangor Darul Ehsan, Subang Jaya, 47500, Malaysia
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Selangor Darul Ehsan, Subang Jaya, 47500, Malaysia
| | - Sin-Yeang Teow
- Department of Biology, College of Science, Mathematics, and Technology, Wenzhou-Kean University, Daxue Road, Ouhai, Wenzhou, 325060, Zhejiang Province, China.
- Wenzhou Municipal Key Laboratory for Applied Biomedical and Biopharmaceutical Informatics, Ouhai, Wenzhou, 325060, Zhejiang Province, China.
- Zhejiang Bioinformatics International Science and Technology Cooperation Center, Ouhai, Wenzhou, 325060, Zhejiang Province, China.
- Dorothy and George Hennings College of Science, Mathematics and Technology, Kean University, 1000 Morris Ave, Union, NJ, 07083, USA.
| |
Collapse
|
2
|
Zhang X, Chen X, Wang A, Wang L, He C, Shi Z, Zhang S, Fu Q, Xu W, Li W, Hu S. Yiqi Jiedu decoction attenuates radiation injury of spermatogenic cells via suppressing IκBα/NF-κB pathway-induced excessive autophagy and apoptosis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116903. [PMID: 37442494 DOI: 10.1016/j.jep.2023.116903] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 06/25/2023] [Accepted: 07/10/2023] [Indexed: 07/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The prescription of Yiqi Jiedu decoction (YQJD) originated from the classic Chinese herbal prescriptions of Danggui Buxue Decoction and Wuzi Yanzong Pill. A previous study has shown that 4 Gy irradiation induced the apoptosis of spermatocytes and revealed autophagosomes in cells exposed to radiation. YQJD decoction has the effect of preventing radiation injury. AIM OF THE STUDY We used spermatocytes (GC-2spd cell line) to investigate the relationship between autophagy and apoptosis of spermatogenic cells after radiation, and the mechanisms of YQJD decoction. MATERIALS AND METHODS Establish an in vitro radiation injury model by irradiating GC-2spd cells with 60Co γ-rays (4 Gy or 8 Gy). Autophagy agonists, autophagy inhibitors and YQJD were used to intervene cells. Cell apoptosis and inflammatory factors were measured. NF-κB localization was observed by immunofluorescence. Autophagy and apoptosis-related proteins and IκBα/NF-κB pathway factors were detected. RESULTS Ionizing radiation promoted the growth of spermatogenic autophagosomes. After radiation, NF-κB was translocated to the nucleus, inflammatory factors were secreted, and IκBα/NF-κB pathway was activated, which promoted autophagy and apoptosis. YQJD decoction can inhibit the phosphorylation of IκBα/NF-κB pathway related factors, regulate the expression of Beclin-1 and Bcl-2 proteins, and inhibit the occurrence of autophagy and apoptosis of irradiated spermatocyte. CONCLUSIONS The research results indicate that ionizing radiation can activate the IκBα/NF-κB signaling pathway in spermatocytes, promote cell autophagy and apoptosis by regulating the expression of Beclin-1 and Bcl-2 factors. The YQJD decoction inhibits the IκBα/NF-κB signaling pathway so as to regulate Beclin-1 and Bcl-2.
Collapse
Affiliation(s)
- Xiaomeng Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China; Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Xiaoying Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - An Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Lei Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Changhao He
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Zhongyu Shi
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Shujing Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Qian Fu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Wenhui Xu
- Beijing Academy of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Wei Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Sumin Hu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
3
|
Abou Shousha S, Osman EM, Baheeg S, Shahine Y. Anti-IL-8 monoclonal antibodies inhibits the autophagic activity and cancer stem cells maintenance within breast cancer tumor microenvironment. Breast Dis 2024; 43:37-49. [PMID: 38552109 PMCID: PMC10977415 DOI: 10.3233/bd-230052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
BACKGROUND Breast cancer tumor microenvironment (TME) is a promising target for immunotherapy. Autophagy, and cancer stem cells (CSCs) maintenance are essential processes involved in tumorigenesis, tumor survival, invasion, and treatment resistance. Overexpression of angiogenic chemokine interleukin-8 (IL-8) in breast cancer TME is associated with oncogenic signaling pathways, increased tumor growth, metastasis, and poor prognosis. OBJECTIVE Thus, we aimed to investigate the possible anti-tumor effect of neutralizing antibodies against IL-8 by evaluating its efficacy on autophagic activity and breast CSC maintenance. METHODS IL-8 monoclonal antibody supplemented tumor tissue culture systems from 15 females undergoing mastectomy were used to evaluate the expression of LC3B as a specific biomarker of autophagy and CD44, CD24 as cell surface markers of breast CSCs using immunofluorescence technique. RESULTS Our results revealed that anti-IL-8 mAb significantly decreased the level of LC3B in the cultured tumor tissues compared to its non-significant decrease in the normal breast tissues.Anti-IL-8 mAb also significantly decreased the CD44 expression in either breast tumors or normal cultured tissues. While it caused a non-significant decrease in CD24 expression in cultured breast tumor tissue and a significant decrease in its expression in the corresponding normal ones. CONCLUSIONS Anti-IL-8 monoclonal antibody exhibits promising immunotherapeutic properties through targeting both autophagy and CSCs maintenance within breast cancer TME.
Collapse
Affiliation(s)
- Seham Abou Shousha
- Immunology and Allergy Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Eman M. Osman
- Immunology and Allergy Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Suzan Baheeg
- Immunology and Allergy Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Yasmine Shahine
- Faculty of Pharmacy, Department of Microbiology & Immunology, Pharos University in Alexandria, Alexandria, Egypt
| |
Collapse
|
4
|
Shen N, Wang L, Wu J, Chen X, Hu F, Su Y. Meta‑analysis of the autophagy‑associated protein LC3 as a prognostic marker in colorectal cancer. Exp Ther Med 2023; 26:492. [PMID: 37753301 PMCID: PMC10518644 DOI: 10.3892/etm.2023.12191] [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: 01/09/2023] [Accepted: 08/02/2023] [Indexed: 09/28/2023] Open
Abstract
Microtubule-associated protein 1 light chain 3 (LC3) is an autophagy-associated gene, which is involved in the progression of a number of human malignancies. Such as Breast Cancer, Liver Cancer, and Lung Cancer. However, the role of LC3 in colorectal cancer (CC) remains to be fully elucidated. Therefore, the prognostic role of LC3 expression in CC was evaluated in the present study, with an emphasis on the clinicopathology and prognosis. Expression of LC3 in CC was examined using PubMed, Cochrane Library, Excerpta Medica Database, China Knowledge Infrastructure and Wanfang Data. Newcastle-Ottawa scale was used to screen the literature quality, and RevMan 5.4 and STATA 14.0 were used for the meta-analysis. A total of 1,689 patients from 10 studies were included in the present meta-analysis. The findings of the present study suggested that increased LC3 expression levels were associated with histological grade [odds ratio (OR)=0.91, 95% confidence interval (CI) (0.47, 1.77), P<0.001] and TNM stage [OR=0.91, 95% CI (0.47, 1.77), P<0.001], but were not associated with sex [OR=1.14, 95% CI (0.90, 1.51)], age [OR=0.89, 95% CI (0.67, 1.20)], tumor size [OR=0.78, 95% CI (0.30, 2.34)], histological grade [OR=0.82, 95% CI (0.43, 1.95)] and lymph node metastasis [OR=2.05, 95% CI (1.19, 3.60)] in CC. In addition, the increased expression of LC3 was revealed to be a prognostic factor for the overall survival of patients with CC. In conclusion, the autophagy-associated protein LC3 may be a prognostic indicator of human CC.
Collapse
Affiliation(s)
- Ning Shen
- Department of Oncology, Zhangqiu People's Hospital, Jinan, Shandong 250200, P.R. China
| | - Lijuan Wang
- Blood Purification Center, Suzhou Science and Technology Town Hospital, Suzhou, Jiangsu 215153, P.R. China
| | - Jingjing Wu
- Department of Oncology, Zhangqiu People's Hospital, Jinan, Shandong 250200, P.R. China
| | - Xuefang Chen
- Blood Purification Center, Suzhou Science and Technology Town Hospital, Suzhou, Jiangsu 215153, P.R. China
| | - Fengchao Hu
- Department of Oncology, Zhangqiu People's Hospital, Jinan, Shandong 250200, P.R. China
| | - Yi Su
- Quality Management Office, Zhangqiu People's Hospital, Jinan, Shandong 250200, P.R. China
| |
Collapse
|
5
|
Rajan PK, Udoh UAS, Nakafuku Y, Pierre SV, Sanabria J. Normalization of the ATP1A1 Signalosome Rescinds Epigenetic Modifications and Induces Cell Autophagy in Hepatocellular Carcinoma. Cells 2023; 12:2367. [PMID: 37830582 PMCID: PMC10572209 DOI: 10.3390/cells12192367] [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: 08/10/2023] [Revised: 09/22/2023] [Accepted: 09/23/2023] [Indexed: 10/14/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related death worldwide. In metabolic dysfunction-associated steatohepatitis (MASH)-related HCC, cellular redox imbalance from metabolic disturbances leads to dysregulation of the α1-subunit of the Na/K-ATPase (ATP1A1) signalosome. We have recently reported that the normalization of this pathway exhibited tumor suppressor activity in MASH-HCC. We hypothesized that dysregulated signaling from the ATP1A1, mediated by cellular metabolic stress, promotes aberrant epigenetic modifications including abnormal post-translational histone modifications and dysfunctional autophagic activity, leading to HCC development and progression. Increased H3K9 acetylation (H3K9ac) and H3K9 tri-methylation (H3K9me3) were observed in human HCC cell lines, HCC-xenograft and MASH-HCC mouse models, and epigenetic changes were associated with decreased cell autophagy in HCC cell lines. Inhibition of the pro-autophagic transcription factor FoxO1 was associated with elevated protein carbonylation and decreased levels of reduced glutathione (GSH). In contrast, normalization of the ATP1A1 signaling significantly decreased H3K9ac and H3K9me3, in vitro and in vivo, with concomitant nuclear localization of FoxO1, heightening cell autophagy and cancer-cell apoptotic activities in treated HCC cell lines. Our results showed the critical role of the ATP1A1 signalosome in HCC development and progression through epigenetic modifications and impaired cell autophagy activity, highlighting the importance of the ATP1A1 pathway as a potential therapeutic target for HCC.
Collapse
Affiliation(s)
- Pradeep Kumar Rajan
- Department of Surgery, Marshall Institute for Interdisciplinary Research, Marshall University School of Medicine, Huntington, WV 25701, USA; (P.K.R.); (U.-A.S.U.); (Y.N.); (S.V.P.)
| | - Utibe-Abasi S. Udoh
- Department of Surgery, Marshall Institute for Interdisciplinary Research, Marshall University School of Medicine, Huntington, WV 25701, USA; (P.K.R.); (U.-A.S.U.); (Y.N.); (S.V.P.)
| | - Yuto Nakafuku
- Department of Surgery, Marshall Institute for Interdisciplinary Research, Marshall University School of Medicine, Huntington, WV 25701, USA; (P.K.R.); (U.-A.S.U.); (Y.N.); (S.V.P.)
| | - Sandrine V. Pierre
- Department of Surgery, Marshall Institute for Interdisciplinary Research, Marshall University School of Medicine, Huntington, WV 25701, USA; (P.K.R.); (U.-A.S.U.); (Y.N.); (S.V.P.)
| | - Juan Sanabria
- Department of Surgery, Marshall Institute for Interdisciplinary Research, Marshall University School of Medicine, Huntington, WV 25701, USA; (P.K.R.); (U.-A.S.U.); (Y.N.); (S.V.P.)
- Department of Nutrition and Metabolomic Core Facility, Case Western Reserve University School of Medicine, Cleveland, OH 44100, USA
| |
Collapse
|
6
|
Ugonabo O, Udoh UAS, Rajan PK, Reeves H, Arcand C, Nakafuku Y, Joshi T, Finley R, Pierre SV, Sanabria JR. The Current Status of the Liver Liquid Biopsy in MASH Related HCC: Overview and Future Directions. Biomolecules 2023; 13:1369. [PMID: 37759769 PMCID: PMC10526956 DOI: 10.3390/biom13091369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/03/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is one of the major risk factors for chronic liver disease and hepatocellular carcinoma (HCC). The incidence of MASH in Western countries continues to rise, driving HCC as the third cause of cancer-related death worldwide. HCC has become a major global health challenge, partly from the obesity epidemic promoting metabolic cellular disturbances but also from the paucity of biomarkers for its early detection. Over 50% of HCC cases are clinically present at a late stage, where curative measures are no longer beneficial. Currently, there is a paucity of both specific and sensitive biological markers for the early-stage detection of HCC. The search for biological markers in the diagnosis of early HCC in high-risk populations is intense. We described the potential role of surrogates for a liver biopsy in the screening and monitoring of patients at risk for nesting HCC.
Collapse
Affiliation(s)
- Onyinye Ugonabo
- Department of Medicine, Marshall University School of Medicine, Marshall University, Huntington, WV 25701, USA; (O.U.); (T.J.)
| | - Utibe-Abasi Sunday Udoh
- Marshall Institute for Interdisciplinary Research, Marshall University School of Medicine, Huntington, WV 25703, USA; (U.-A.S.U.); (P.K.R.); (Y.N.); (S.V.P.)
- Department of Surgery, Marshall University School of Medicine, Marshall University, Huntington, WV 25701, USA; (H.R.); (C.A.); (R.F.)
| | - Pradeep Kumar Rajan
- Marshall Institute for Interdisciplinary Research, Marshall University School of Medicine, Huntington, WV 25703, USA; (U.-A.S.U.); (P.K.R.); (Y.N.); (S.V.P.)
- Department of Surgery, Marshall University School of Medicine, Marshall University, Huntington, WV 25701, USA; (H.R.); (C.A.); (R.F.)
| | - Heather Reeves
- Department of Surgery, Marshall University School of Medicine, Marshall University, Huntington, WV 25701, USA; (H.R.); (C.A.); (R.F.)
| | - Christina Arcand
- Department of Surgery, Marshall University School of Medicine, Marshall University, Huntington, WV 25701, USA; (H.R.); (C.A.); (R.F.)
| | - Yuto Nakafuku
- Marshall Institute for Interdisciplinary Research, Marshall University School of Medicine, Huntington, WV 25703, USA; (U.-A.S.U.); (P.K.R.); (Y.N.); (S.V.P.)
- Department of Surgery, Marshall University School of Medicine, Marshall University, Huntington, WV 25701, USA; (H.R.); (C.A.); (R.F.)
| | - Tejas Joshi
- Department of Medicine, Marshall University School of Medicine, Marshall University, Huntington, WV 25701, USA; (O.U.); (T.J.)
| | - Rob Finley
- Department of Surgery, Marshall University School of Medicine, Marshall University, Huntington, WV 25701, USA; (H.R.); (C.A.); (R.F.)
| | - Sandrine V. Pierre
- Marshall Institute for Interdisciplinary Research, Marshall University School of Medicine, Huntington, WV 25703, USA; (U.-A.S.U.); (P.K.R.); (Y.N.); (S.V.P.)
| | - Juan Ramon Sanabria
- Marshall Institute for Interdisciplinary Research, Marshall University School of Medicine, Huntington, WV 25703, USA; (U.-A.S.U.); (P.K.R.); (Y.N.); (S.V.P.)
- Department of Surgery, Marshall University School of Medicine, Marshall University, Huntington, WV 25701, USA; (H.R.); (C.A.); (R.F.)
- Department of Nutrition and Metabolomic Core Facility, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
7
|
Li R, Liu X, Deng K, Wang X. M7G methylated core genes (METTL1 and WDR4) and associated RNA risk signatures are associated with prognosis and immune escape in HCC. BMC Med Genomics 2023; 16:179. [PMID: 37528384 PMCID: PMC10394781 DOI: 10.1186/s12920-023-01614-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 07/21/2023] [Indexed: 08/03/2023] Open
Abstract
N7 methylguanosine (m7G) has a crucial role the development of hepatocellular carcinoma (HCC). This study aimed to investigate the impact of the m7G methylation core genes (METTL1 and WDR4) and associated RNA risk signatures on HCC. we found m7G methylation core genes (METTL1 and WDR4) were upregulated in four HCC cell lines, and downregulation of METTL1 and WDR4 attenuated HCC cell proliferation, migration, and invasion. Moreover, METTL1 and WDR4 are upregulated in HCC tissues, and that there is a significant positive correlation between them. METTL1 and WDR4 were identified as independent prognostic markers for HCC by employing overall survival (OS), disease-specific survival (DSS), Progression Free Interval survival (PFI), and univariate/multivariate Cox analyses. We identified 1479 coding RNAs (mRNAs) and 232 long non-coding RNAs (lncRNAs) associated with METTL1 / WDR4 by using weighted coexpression network analysis (WGCNA) and co-clustering analysis. The least absolute shrinkage and selection operator (lasso) were used to constructing mRNA and lncRNA risk signatures associated with the METTL1 / WDR4. These risk were independent poor prognostic factors in HCC. Furthermore, we found that METTL1 / WDR4 expression and mRNA / lncRNA risk scores were closely associated with TP53 mutations. Clinicopathological features correlation results showed that METTL1 / WDR4 expression and mRNA / lncRNA risk score were associated with the stage and invasion depth (T) of HCC. To predict the overall survival of HCC individuals, we constructed a nomogram with METTL1/WDR4 expression, mRNA/lncRNA risk score, and clinicopathological features. In addition, we combined single-cell sequencing datasets and immune escape-related checkpoints to construct an immune escape-related protein-protein interaction(PPI) network. In conclusion, M7G methylated core genes (METTL1 and WDR4) and associated RNA risk signatures are associated with prognosis and immune escape in HCC.
Collapse
Affiliation(s)
- Rui Li
- Jiangnan University Medical Center, WuXi, China
- Wuxi No.2 People's Hospital, WuXi, China
- The Affiliated Wuxi No.2 People's Hospital of Clinical College of Nantong University, WuXi, China
| | | | - Kaiyuan Deng
- Jiangnan University Medical Center, WuXi, China
- Wuxi No.2 People's Hospital, WuXi, China
- The Affiliated Wuxi No.2 People's Hospital of Clinical College of Nantong University, WuXi, China
| | - Xin Wang
- Jiangnan University Medical Center, WuXi, China.
- Wuxi No.2 People's Hospital, WuXi, China.
- The Affiliated Wuxi No.2 People's Hospital of Clinical College of Nantong University, WuXi, China.
| |
Collapse
|
8
|
Huang H, Pan R, Wang S, Guan Y, Zhao Y, Liu X. Current and potential roles of RNA modification-mediated autophagy dysregulation in cancer. Arch Biochem Biophys 2023; 736:109542. [PMID: 36758911 DOI: 10.1016/j.abb.2023.109542] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 01/31/2023] [Accepted: 02/05/2023] [Indexed: 02/09/2023]
Abstract
Autophagy, a cellular lysosomal degradation and survival pathway, supports nutrient recycling and adaptation to metabolic stress and participates in various stages of tumor development, including tumorigenesis, metastasis, and malignant state maintenance. Among the various factors contributing to the dysregulation of autophagy in cancer, RNA modification can regulate autophagy by directly affecting the expression of core autophagy proteins. We propose that autophagy disorder mediated by RNA modification is an important mechanism for cancer development. Therefore, this review mainly discusses the role of RNA modification-mediated autophagy regulation in tumorigenesis. We summarize the molecular basis of autophagy and the core proteins and complexes at different stages of autophagy, especially those involved in cancer development. Moreover, we describe the crosstalk of RNA modification and autophagy and review the recent advances and potential role of the RNA modification/autophagy axis in the development of multiple cancers. Furthermore, the dual role of the RNA modification/autophagy axis in cancer drug resistance is discussed. A comprehensive understanding and extensive exploration of the molecular crosstalk of RNA modifications with autophagy will provide important insights into tumor pathophysiology and provide more options for cancer therapeutic strategies.
Collapse
Affiliation(s)
- Hua Huang
- Center of Excellence for Environmental Safety and Biological Effects, Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Faculty of Environment and Life, Beijing University of Technology, Beijing, 100124, China
| | - Ruining Pan
- Center of Excellence for Environmental Safety and Biological Effects, Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Faculty of Environment and Life, Beijing University of Technology, Beijing, 100124, China
| | - Sijia Wang
- Center of Excellence for Environmental Safety and Biological Effects, Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Faculty of Environment and Life, Beijing University of Technology, Beijing, 100124, China
| | - Yifei Guan
- Center of Excellence for Environmental Safety and Biological Effects, Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Faculty of Environment and Life, Beijing University of Technology, Beijing, 100124, China
| | - Yue Zhao
- Intensive Care Unit, Beijing Tsinghua Changgung Hospital, Beijing, 102218, China.
| | - Xinhui Liu
- Center of Excellence for Environmental Safety and Biological Effects, Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Faculty of Environment and Life, Beijing University of Technology, Beijing, 100124, China.
| |
Collapse
|
9
|
Wei Y, Li Y, Chen Y, Liu P, Huang S, Zhang Y, Sun Y, Wu Z, Hu M, Wu Q, Wu H, Liu F, She T, Ning Z. ALDH1: A potential therapeutic target for cancer stem cells in solid tumors. Front Oncol 2022; 12:1026278. [PMID: 36387165 PMCID: PMC9650078 DOI: 10.3389/fonc.2022.1026278] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/12/2022] [Indexed: 12/02/2022] Open
Abstract
Solid tumors can be divided into benign solid tumors and solid malignant tumors in the academic community, among which malignant solid tumors are called cancers. Cancer is the second leading cause of death in the world, and the global incidence of cancer is increasing yearly New cancer patients in China are always the first. After the concept of stem cells was introduced in the tumor community, the CSC markers represented by ALDH1 have been widely studied due to their strong CSC cell characteristics and potential to be the driving force of tumor metastasis. In the research results in the past five years, it has been found that ALDH1 is highly expressed in various solid cancers such as breast cancer, lung cancer, colorectal cancer, liver cancer, gastric cancer, cervical cancer, esophageal cancer, ovarian cancer, head,and neck cancer. ALDH1 can activate and transform various pathways (such as the USP28/MYC signaling pathway, ALDH1A1/HIF-1α/VEGF axis, wnt/β-catenin signaling pathway), as well as change the intracellular pH value to promote formation and maintenance, resulting in drug resistance in tumors. By targeting and inhibiting ALDH1 in tumor stem cells, it can enhance the sensitivity of drugs and inhibit the proliferation, differentiation, and metastasis of solid tumor stem cells to some extent. This review discusses the relationship and pathway of ALDH1 with various solid tumors. It proposes that ALDH1 may serve as a diagnosis and therapeutic target for CSC, providing new insights and new strategies for reliable tumor treatment.
Collapse
Affiliation(s)
- Yaolu Wei
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Yan Li
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Yenan Chen
- Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Pei Liu
- Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Sheng Huang
- Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Yuping Zhang
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Yanling Sun
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Zhe Wu
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Meichun Hu
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Qian Wu
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Hongnian Wu
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Fuxing Liu
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
- *Correspondence: Fuxing Liu, ; Tonghui She, ; Zhifeng Ning,
| | - Tonghui She
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
- *Correspondence: Fuxing Liu, ; Tonghui She, ; Zhifeng Ning,
| | - Zhifeng Ning
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
- *Correspondence: Fuxing Liu, ; Tonghui She, ; Zhifeng Ning,
| |
Collapse
|
10
|
Yang J, Guo W, Lu M. Recent Perspectives on the Mechanism of Recurrence After Ablation of Hepatocellular Carcinoma: A Mini-Review. Front Oncol 2022; 12:895678. [PMID: 36081558 PMCID: PMC9445307 DOI: 10.3389/fonc.2022.895678] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 05/16/2022] [Indexed: 11/28/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignant tumors. Hepatectomy, liver transplantation, and ablation are the three radical treatments for early-stage hepatocellular carcinoma (ESHCC), but not all patients are fit for or can tolerate surgery; moreover, liver donors are limited. Therefore, ablation plays an important role in the treatment of ESHCC. However, some studies have shown that ablation has a higher local recurrence (LR) rate than hepatectomy and liver transplantation. The specific mechanism is unknown. The latest perspectives on the mechanism of recurrence after ablation of HCC were described and summarized. In this review, we discussed the possible mechanisms of recurrence after ablation of HCC, including epithelial–mesenchymal transition (EMT), activating autophagy, changes in non-coding RNA, and changes in the tumor microenvironment. A systematic and comprehensive understanding of the mechanism will contribute to the research and development of related treatment, combined with ablation to improve the therapeutic effect in patients with ESHCC.
Collapse
Affiliation(s)
- Jianquan Yang
- The School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Department of Ultrasound Medical Center, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Wen Guo
- Institute of Materia Medica, North Sichuan Medical College, Nanchong, China
| | - Man Lu
- The School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Department of Ultrasound Medical Center, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- *Correspondence: Man Lu,
| |
Collapse
|
11
|
Xie Y, Liu Y, Sun J, Zheng L. Synthesis of mitochondria-targeted ferulic acid amide derivatives with antioxidant, anti-inflammatory activities and inducing mitophagy. Bioorg Chem 2022; 127:106037. [PMID: 35863132 DOI: 10.1016/j.bioorg.2022.106037] [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: 05/05/2022] [Revised: 07/12/2022] [Accepted: 07/14/2022] [Indexed: 11/18/2022]
Abstract
The seventeen ferulic acid amide derivatives were synthesized by coupling mitochondrial carrier coumarin-3-carboxamide with acrylic acids. The results of cellular antioxidant activity and inhibitory effects on NO production against LPS-stimulated RAW264.7 macrophages indicated four compounds (8c, 8d, 9c, 9d) showed the higher dual-activities of antioxidant and anti-inflammatory. The structure-activity relationship was deduced. In regard to mechanism research, the most potent compound 8d which mainly distributed in mitochondria suppressed the secretion of inflammatory cytokines IL-6 and TNF-α, enhancing mitophagy to alleviate inflammatory response. Besides, the dual-activities were diminished by removal of coumarin carrier in 8d, suggesting the enrichment in mitochondria might be important for activities. This study showed that development of mitochondria-targeted antioxidants could be a feasible strategy to resist inflammation.
Collapse
Affiliation(s)
- Yu Xie
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Yongpeng Liu
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Jing Sun
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Lifang Zheng
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
12
|
An Experimental Study Reveals the Protective Effect of Autophagy against Realgar-Induced Liver Injury via Suppressing ROS-Mediated NLRP3 Inflammasome Pathway. Int J Mol Sci 2022; 23:ijms23105697. [PMID: 35628508 PMCID: PMC9145910 DOI: 10.3390/ijms23105697] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/12/2022] [Accepted: 05/18/2022] [Indexed: 12/25/2022] Open
Abstract
Realgar, a poisonous traditional Chinese medicine, has been shown to cause liver injury when used for long periods or overdoses. However, the underlying molecular mechanisms and therapeutic targets have not been fully elucidated. The aim of this study is to explore the role of autophagy in sub-chronic realgar exposure-induced liver injury. Here, the liver injury model was established by continuously administrating mice with 1.35 g/kg realgar for 8 weeks. 3-methyladenine (3-MA) and rapamycin (RAPA) were used to regulate autophagy. The results showed that realgar induced abnormal changes in liver function, pathological morphology, expression of inflammatory cytokines, and upregulated NLRP3 inflammasome pathway in mouse livers. RAPA treatment (an inducer of autophagy) significantly improved realgar-induced liver injury and NLRP3 inflammasome activation, while 3-MA (an inhibitor of autophagy) aggravated the realgar-induced liver injury and NLRP3 inflammasome activation. Furthermore, we found that realgar-induced NLRP3 inflammasome activation in mouse livers is mediated by ROS. RAPA eliminates excessive ROS, inhibits NF-κB nuclear translocation and down-regulates the TXNIP/NLRP3 axis, consequently suppressing ROS-mediated NLRP3 inflammasome activation, which may be the underlying mechanism of the protective effect of autophagy on realgar-induced liver injury. In conclusion, the results of this study suggest that autophagy alleviates realgar-induced liver injury by inhibiting ROS-mediated NLRP3 inflammasome activation. Autophagy may represent a therapeutic target in modulating realgar-induced liver injury.
Collapse
|
13
|
Ning Z, Lan J, Jiang X, Zhong G, Zhang H, Wan F, Wu S, Tang Z, Bilal RM, Hu L, Huang R. Arsenic trioxide-induced autophagy affected the antioxidant capacity and apoptosis rate of chicken hepatocytes. Chem Biol Interact 2022; 354:109821. [PMID: 35051378 DOI: 10.1016/j.cbi.2022.109821] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 12/20/2021] [Accepted: 01/13/2022] [Indexed: 12/19/2022]
Abstract
Arsenic has recently received widespread attention due to its high toxicological effects on multiple animals; however, the mechanism underlying this toxicity is unclear. We investigated the damaging effects of arsenic trioxide (ATO) on hepatocytes and the effects of regulating autophagy on the hepatocyte damage induced by ATO exposure. First, we investigated the effects of ATO exposure (0, 0.6, 1.2, 2.4, and 4.8 μM) on the biochemical function and autophagy of chicken hepatocytes. The findings showed that as the concentration of ATO increased, the lactate dehydrogenase (LDH) concentration increased, more autophagosomes were observed via transmission electron microscopy (TEM), and the gene and protein expression levels of P62, LC3Ⅱ, and Beclin1 increased. Adding N-acetyl-l-cystine (NAC, 1 mM) attenuated autophagy and the hepatocyte damage induced by ATO. Then, we used rapamycin (Rapa) and 3-methylpurine (3-MA) to regulate the autophagy induced by exposure to 4.8 μM ATO and observed changes in the antioxidant capacity and apoptosis rate of chicken hepatocytes. Induction of autophagy reduced ATO-induced hepatocyte apoptosis but caused no significant effect on oxidative stress in chicken hepatocytes. Inhibition of autophagy exacerbated ATO-induced hepatocyte oxidative stress and apoptosis. These findings demonstrate that autophagy plays an important role in ATO-induced cell damage.
Collapse
Affiliation(s)
- Zhijun Ning
- College of Veterinary Medicine, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, China.
| | - Juan Lan
- College of Veterinary Medicine, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, China.
| | - Xuanxuan Jiang
- College of Veterinary Medicine, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, China.
| | - Gaolong Zhong
- College of Veterinary Medicine, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, China.
| | - Hui Zhang
- College of Veterinary Medicine, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, China.
| | - Fang Wan
- College of Veterinary Medicine, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, China.
| | - Shaofeng Wu
- College of Veterinary Medicine, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, China.
| | - Zhaoxin Tang
- College of Veterinary Medicine, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, China.
| | - Rana Muhammad Bilal
- University College of Veterinary & Animal Sciences, Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan.
| | - Lianmei Hu
- College of Veterinary Medicine, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, China.
| | - Riming Huang
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
14
|
Kim MJ, Choi B, Kim JY, Min Y, Kwon DH, Son J, Lee JS, Lee JS, Chun E, Lee KY. USP8 regulates liver cancer progression via the inhibition of TRAF6-mediated signal for NF-κB activation and autophagy induction by TLR4. Transl Oncol 2022; 15:101250. [PMID: 34688043 PMCID: PMC8546492 DOI: 10.1016/j.tranon.2021.101250] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 12/12/2022] Open
Abstract
Herein, we aimed to elucidate the molecular and cellular mechanism in which ubiquitin-specific protease 8 (USP8) is implicated in liver cancer progression via TRAF6-mediated signal. USP8 induces the deubiquitination of TRAF6, TAB2, TAK1, p62, and BECN1, which are pivotal roles for NF-κB activation and autophagy induction. Notably, the LIHC patient with low USP8 mRNA expression showed markedly shorter survival time, whereas there was no significant difference in the other 18-human cancers. Importantly, the TCGA data analysis on LIHC and transcriptome analysis on the USP8 knockout (USP8KO) SK-HEP-1 cells revealed a significant correlation between USP8 and TRAF6, TAB2, TAK1, p62, and BECN1, and enhanced NF-κB-dependent and autophagy-related cancer progression/metastasis-related genes in response to LPS stimulation. Furthermore, USP8KO SK-HEP-1 cells showed an increase in cancer migration and invasion by TLR4 stimulation, and a marked increase of tumorigenicity and metastasis in xenografted NSG mice. The results demonstrate that USP8 is negatively implicated in the LIHC progression through the regulation of TRAF6-mediated signal for the activation of NF-κB activation and autophagy induction. Our findings provide useful insight into the LIHC pathogenesis of cancer progression.
Collapse
Affiliation(s)
- Mi-Jeong Kim
- Department of Immunology and Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Bongkum Choi
- Department of Medicine, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Ji Young Kim
- Department of Immunology and Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Yoon Min
- Department of Immunology and Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Do Hee Kwon
- Department of Immunology and Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Juhee Son
- Department of Immunology and Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Ji Su Lee
- Department of Immunology and Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Joo Sang Lee
- Department of Precision medicine, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Eunyoung Chun
- CHA Vaccine Institute, 560 Dunchon-daero, Jungwon-gu, Seongnam-si, Gyeonggi-do 13230, Republic of Korea.
| | - Ki-Young Lee
- Department of Immunology and Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea; Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Samsung Medical Center, Sungkyunkwan University, Seoul, Republic of Korea.
| |
Collapse
|
15
|
Sun S, Zhang Y, Xu W, Wang B, Chen J. Mitochondrial dysfunction is involved in the cellular activity inhibition by eleutheroside B in SMMC-7721 and HeLa cells. Hum Exp Toxicol 2022; 41:9603271221089006. [PMID: 35580997 DOI: 10.1177/09603271221089006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Eleutheroside B, also known as syringin, has been shown to have various pharmacological activities such as anti-inflammatory, anti-irradiation and antidepressant, but there are few studies on its anti-cancer activity. Its anti-tumor effect on SMMC-7721 cells has not been revealed. Moreover, whether it induces autophagy is still unclear. Thus, the present study investigated whether Eleutheroside B induces apoptosis, autophagy and cellular motility in SMMC-7721 cells and HeLa cells, and explored the underlying molecular mechanisms. SMMC-7721 cells and HeLa cells treated with Eleutheroside B and cell viability measured by MTT assay and trypan blue dye exclusion assay. Apoptosis checked by flow cytometry combined, fluorescent staining. Apoptotic signal proteins and autophagy proteins were checked by Western blot. This study showed that Eleutheroside B inhibited the cell proliferation and blocked cell cycle, migration and invasion as well. Moreover, Eleutheroside B induced apoptosis in SMMC-7721 cells and HeLa cells. It upregulated Bax expression, while simultaneously decreasing Bcl-2 expression. Further elucidation of the mechanism revealed that Eleutheroside B induced mitochondrial dysfunction, with mitochondrial membrane potential collapse and cytochrome c release, suggesting that Eleutheroside B induced apoptosis by triggering mitochondrial pathway. Most importantly, Eleutheroside B could induce autophagy in SMMC-7721 cells and HeLa cells. Taken together, these results suggested Eleutheroside B is a potential therapeutic candidate for HCC and Human cervical cancer.
Collapse
Affiliation(s)
- Shangfan Sun
- School of Basic Medical Sciences, 128790North China University of Science and Technology, China.,Hebei Key Laboratory for Chronic Diseases, 128790North China University of Science and Technology, China
| | - Yi Zhang
- School of Basic Medical Sciences, 519884Xiamen Medical College, China
| | - Weili Xu
- College of Life Sciences, 128790North China University of Science and Technology, China
| | - Bin Wang
- School of Basic Medical Sciences, 128790North China University of Science and Technology, China
| | - Jing Chen
- College of Life Sciences, 128790North China University of Science and Technology, China
| |
Collapse
|
16
|
Elsayed MS, Abu-Elsaad NM, Nader MA. The NLRP3 inhibitor dapansutrile attenuates folic acid induced nephrotoxicity via inhibiting inflammasome/caspase-1/IL axis and regulating autophagy/proliferation. Life Sci 2021; 285:119974. [PMID: 34560082 DOI: 10.1016/j.lfs.2021.119974] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/15/2021] [Accepted: 09/17/2021] [Indexed: 11/26/2022]
Abstract
AIMS Chemical renal toxicity is common and has limited therapeutic interventions. The NLRP3 inhibitor dapansutrile (DAPA) undergoes clinical phase II trials and it shows promising beneficial effects in various inflammatory diseases. The current study aims at evaluating the effect of DAPA on folic acid (FA) induced acute kidney injury (AKI) and its possible transition to chronic injury. MATERIALS AND METHODS Two treatment protocols were studied depending on DAPA injection timing. A prophylactic protocol involving the injection of DAPA (0.2 mg/kg) daily for seven days before FA challenge and a therapeutic protocol where DAPA was injected after FA. Each protocol included four groups of rats: control group, DAPA group, FA group and DAPA+FA group. Serum creatinine, urea and uric acid were measured. Also, kidney injury, necrosis and fibrosis percentage in addition to infiltration of CD68 positive cells were evaluated. Activation markers of inflammasome and the expression of Ki-67 and LC-3 were measured. KEY FINDINGS Results showed an improvement in renal tissue integrity and a significant decrease in kidney function biomarkers, caspase-1, IL-1β and IL-18 by DAPA injection (p < 0.05). In addition, DAPA decreased the proliferation marker Ki-67 and the autophagic marker LC-3 (p < 0.01). SIGNIFICANCE DAPA potentially alleviates FA induced nephrotoxicity through targeting inflammasome/caspase-1/IL axis. Moreover, it shows a regulatory effect on renal regeneration and autophagy.
Collapse
Affiliation(s)
- Mohamed S Elsayed
- Pharmacology and Toxicology Dep., Faculty of Pharmacy, Mansoura University, Egypt
| | - Nashwa M Abu-Elsaad
- Pharmacology and Toxicology Dep., Faculty of Pharmacy, Mansoura University, Egypt.
| | - Manar A Nader
- Pharmacology and Toxicology Dep., Faculty of Pharmacy, Mansoura University, Egypt
| |
Collapse
|
17
|
Deng X, Zhao F, Zhao D, Zhang Q, Zhu Y, Chen Q, Qiang L, Xie N, Ma J, Pan X, Wu Y, Guan L, Xie Y. Oxymatrine promotes hypertrophic scar repair through reduced human scar fibroblast viability, collagen and induced apoptosis via autophagy inhibition. Int Wound J 2021; 19:1221-1231. [PMID: 34749441 PMCID: PMC9284648 DOI: 10.1111/iwj.13717] [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: 07/31/2021] [Revised: 10/20/2021] [Accepted: 10/26/2021] [Indexed: 12/21/2022] Open
Abstract
Scars are common complications of burns and trauma, resulting in mental trauma, physical pain, and a heavy financial burden for patients. Specific and effective anti‐scarring drugs are lacking in clinical practice. Phytochemicals are easily accessible, low in toxicity, and have various biological and pharmacological properties. Oxymatrine is a phytochemical that regulates autophagy networks. Autophagy is closely related to the maintenance, activity, differentiation, and life‐death of skin fibroblasts during wound repair, which results in pathological scars. We hypothesised that oxymatrine may promote hypertrophic scar repair by inhibiting fibroblast autophagy. In vitro studies showed that inhibition of autophagy by oxymatrine decreased viability and collagen metabolism, and increased apoptosis of human scar fibroblasts (HSFs). In vivo studies showed that inhibition of autophagy by oxymatrine promoted scar repair, resulting in a significantly improved final outcome of the hypertrophic scars, a smaller scar area, decreased epidermal and dermal thickness, and a significant downregulation of CK10, P63, collagen I, α‐SMA, and TGF‐β1. In summary, oxymatrine promoted hypertrophic scar repair by decreasing HSF viability and collagen, and inducing apoptosis via autophagy inhibition. This study provides a new perspective on the mechanism of hypertrophic burn scar formation, as well as key scientific data for the application of the phytochemical oxymatrine as a new method for the prevention and treatment of hypertrophic scars.
Collapse
Affiliation(s)
- Xingwang Deng
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China.,Department of Burns and Plastic Surgery, The First People's Hospital of Shizuishan, Shizuishan, China
| | - Fang Zhao
- Tissue Organ Bank & Tissue Engineering Centre, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Dan Zhao
- Tissue Organ Bank & Tissue Engineering Centre, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Qing Zhang
- Tissue Organ Bank & Tissue Engineering Centre, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Yongzhao Zhu
- Tissue Organ Bank & Tissue Engineering Centre, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Qian Chen
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China.,Department of Burns and Plastic Surgery, Xinyang Central Hospital, Xinyang, China
| | - Lijuan Qiang
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China.,Department of Burns and Plastic Surgery, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, China
| | - Nan Xie
- Beijing Engineering Lab for Cell Therapy, Beijing, China
| | - Jiaxiang Ma
- Tissue Organ Bank & Tissue Engineering Centre, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Xiaoliang Pan
- Tissue Organ Bank & Tissue Engineering Centre, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Yinsheng Wu
- Department of Burns and Plastic Surgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Lifeng Guan
- Department of Burns and Plastic Surgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Yan Xie
- Tissue Organ Bank & Tissue Engineering Centre, General Hospital of Ningxia Medical University, Yinchuan, China.,School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
18
|
Niture S, Lin M, Rios-Colon L, Qi Q, Moore JT, Kumar D. Emerging Roles of Impaired Autophagy in Fatty Liver Disease and Hepatocellular Carcinoma. Int J Hepatol 2021; 2021:6675762. [PMID: 33976943 PMCID: PMC8083829 DOI: 10.1155/2021/6675762] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 03/16/2021] [Accepted: 04/06/2021] [Indexed: 02/07/2023] Open
Abstract
Autophagy is a conserved catabolic process that eliminates dysfunctional cytosolic biomolecules through vacuole-mediated sequestration and lysosomal degradation. Although the molecular mechanisms that regulate autophagy are not fully understood, recent work indicates that dysfunctional/impaired autophagic functions are associated with the development and progression of nonalcoholic fatty liver disease (NAFLD), alcoholic fatty liver disease (AFLD), and hepatocellular carcinoma (HCC). Autophagy prevents NAFLD and AFLD progression through enhanced lipid catabolism and decreasing hepatic steatosis, which is characterized by the accumulation of triglycerides and increased inflammation. However, as both diseases progress, autophagy can become impaired leading to exacerbation of both pathological conditions and progression into HCC. Due to the significance of impaired autophagy in these diseases, there is increased interest in studying pathways and targets involved in maintaining efficient autophagic functions as potential therapeutic targets. In this review, we summarize how impaired autophagy affects liver function and contributes to NAFLD, AFLD, and HCC progression. We will also explore how recent discoveries could provide novel therapeutic opportunities to effectively treat these diseases.
Collapse
Affiliation(s)
- Suryakant Niture
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, NC 27707, USA
| | - Minghui Lin
- The Fourth People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, China 750021
| | - Leslimar Rios-Colon
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, NC 27707, USA
| | - Qi Qi
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, NC 27707, USA
| | - John T. Moore
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, NC 27707, USA
| | - Deepak Kumar
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, NC 27707, USA
| |
Collapse
|
19
|
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
|
20
|
Hou J, Zhao L, Tang H, He X, Ye G, Shi F, Kang M, Chen H, Li Y. Silver Nanoparticles Induced Oxidative Stress and Mitochondrial Injuries Mediated Autophagy in HC11 Cells Through Akt/AMPK/mTOR Pathway. Biol Trace Elem Res 2021; 199:1062-1073. [PMID: 32666434 DOI: 10.1007/s12011-020-02212-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 05/19/2020] [Indexed: 12/14/2022]
Abstract
Silver nanoparticles (AgNPs) are widely used in industrial products, and they have good antibacterial properties, with potential for prevention and treatment of cow mastitis. However, concerns exist about the cytotoxicity of AgNPs. Thus, we have studied the role of autophagy in AgNP-induced cytotoxicity in mouse HC11 mammary epithelium cells. We found that AgNPs injured HC11 cells, with release of lactate dehydrogenase (LDH). AgNPs also induced autophagy in HC11 cells, which was associated with oxidative stress, as indicated by increased reactive oxygen species (ROS) and increased expression of hemoxygenase-1(HO-1) and Nrf2. Mitochondria were altered by AgNPs: mitochondrial membrane potential (MMP) was decreased and the expression of PINK1 and Parkin was increased. AgNPs also increased the expression of p-AMPK and decreased the expression of p-Akt and p-mTOR. The addition of 3-methyl adenine inhibited autophagy and enhanced the cytotoxicity of AgNPs, indicating that autophagy is protective against AgNP-induced cell death. In summary, AgNPs induced protective autophagy in HC11 cells via the Akt/AMPK/mTOR pathway, associated with cellular oxidative stress and mitochondrial alterations. Our research confirms that AgNPs may damage the breast tissue in clinical applications and should be used with caution. Further research is necessary to clarify whether the damage caused by AgNPs will affect the lactation function of the mammary glands and possible residues in milk.
Collapse
Affiliation(s)
- Jin Hou
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu, 611130, Sichuan, China
| | - Ling Zhao
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu, 611130, Sichuan, China
| | - Huaqiao Tang
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu, 611130, Sichuan, China
| | - Xiaoli He
- College of Science, Sichuan Agricultural University, 211 Huimin Road, Chengdu, 611130, Sichuan, China
| | - Gang Ye
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu, 611130, Sichuan, China
| | - Fei Shi
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu, 611130, Sichuan, China
| | - Min Kang
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu, 611130, Sichuan, China
| | - Helin Chen
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu, 611130, Sichuan, China
| | - Yinglun Li
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu, 611130, Sichuan, China.
| |
Collapse
|
21
|
Wang K, Zheng J. Knockdown of BAG3 synergizes with olaparib to kill ovarian cancer cells via repressing autophagy. J Investig Med 2020; 69:jim-2020-001602. [PMID: 33361105 DOI: 10.1136/jim-2020-001602] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2020] [Indexed: 11/04/2022]
Abstract
This study aimed at expounding the synergistic effect of Bcl-2-associated athanogene 3 (BAG3) knockdown and poly ADP-ribose polymerase (PARP) inhibitor on ovarian cancer (OC) cells and the potential mechanism. Short hairpin RNA (shRNA) targeting BAG3 (sh-BAG3) was transfected into SK-OV-3 (SKOV-3 ;SKOV3) and A2780 cells, and western blot assay was used to detect transfection efficiency. Cell proliferation and apoptosis were detected by the cell counting kit-8 method, 5-Bromodeoxyuridine (BrdU) experiment and flow cytometry analysis, respectively. The expressions of apoptosis-related proteins Bax and Bcl-2, as well as the expressions of autophagy-related proteins LC3-I, LC3-II and Beclin-1, were examined by western blot assay. Additionally, the cells were treated with autophagy activator rapamycin to investigate whether the tumor-suppressive function of BAG3 knockdown+PARP inhibitor was dependent on autophagy. In this work, we demonstrated that BAG3 knockdown further sensitized OC cells to olaparib treatment, reducing cellular viability and promoting apoptosis. Both sh-BAG3 and olaparib decreased the expression of Beclin-1 and the LC3-Ⅱ:LC3-I ratio, and their synergism further inhibited the process of autophagy. However, the aforementionede effects were reversed after the cells were treated with rapamycin. Based on these results, we concluded that BAG3 knockdown synergizes with olaparib to kill OC cells in vitro by repressing autophagy.
Collapse
Affiliation(s)
- Kexin Wang
- Obstetrics and Gynecology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Jianhua Zheng
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
22
|
The Role of Autophagy in Liver Cancer: Crosstalk in Signaling Pathways and Potential Therapeutic Targets. Pharmaceuticals (Basel) 2020; 13:ph13120432. [PMID: 33260729 PMCID: PMC7760785 DOI: 10.3390/ph13120432] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 11/26/2020] [Accepted: 11/26/2020] [Indexed: 02/06/2023] Open
Abstract
Autophagy is an evolutionarily conserved lysosomal-dependent pathway for degrading cytoplasmic proteins, macromolecules, and organelles. Autophagy-related genes (Atgs) are the core molecular machinery in the control of autophagy, and several major functional groups of Atgs coordinate the entire autophagic process. Autophagy plays a dual role in liver cancer development via several critical signaling pathways, including the PI3K-AKT-mTOR, AMPK-mTOR, EGF, MAPK, Wnt/β-catenin, p53, and NF-κB pathways. Here, we review the signaling pathways involved in the cross-talk between autophagy and hepatocellular carcinoma (HCC) and analyze the status of the development of novel HCC therapy by targeting the core molecular machinery of autophagy as well as the key signaling pathways. The induction or the inhibition of autophagy by the modulation of signaling pathways can confer therapeutic benefits to patients. Understanding the molecular mechanisms underlying the cross-link of autophagy and HCC may extend to translational studies that may ultimately lead to novel therapy and regimen formation in HCC treatment.
Collapse
|
23
|
Yang W, Niu L, Zhao X, Duan L, Li Y, Wang X, Zhang Y, Zhou W, Liu J, Zhao Q, Han Y, Fan D, Hong L. Development and validation of a survival model based on autophagy-associated genes for predicting prognosis of hepatocellular carcinoma. Am J Transl Res 2020; 12:6705-6722. [PMID: 33194067 PMCID: PMC7653605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 09/24/2020] [Indexed: 06/11/2023]
Abstract
OBJECTIVE This study aimed to identify the novel prognostic gene signature based on autophagy-associated genes (ARGs) in hepatocellular carcinoma (HCC). METHODS The RNA sequencing data and clinical information of HCC and normal tissues were obtained from The Cancer Genome Atlas (TCGA) database. The differentially expressed ARGs were screened by the Wilcoxon signed-rank test. Cox regression analysis and Lasso regression analysis were performed to screen the ARGs and establish the prognostic prediction model. Kaplan-Meier and receiver operating characteristic (ROC) curves were both used to evaluate the accuracy of the model. GSE14520 dataset (testing cohort) was used to validate the prognostic risk model in TCGA. A clinical nomogram was established to predict the survival rate of HCC patients. RESULTS Totally 27 differentially expressed ARGs were identified. Three OS-related ARGs (SQSTM1, HSPB8, and BIRC5) were identified via the Cox regression and Lasso regression analyses. Based on these three ARGs, a prognostic prediction model was constructed. HCC patients with high risk score present poorer prognosis than those with low risk score both in TCGA cohort (P=4.478e-04) and testing cohort (P=1.274e-03). Moreover, the risk score curve shows a well feasibility in predicting the patients' survival both in TCGA and GEO cohort with the area under the ROC curve (AUC) of 0.756 and 0.672, respectively. Besides, the calibration curves and C-index indicated that the clinical nomogram performs well to predict survival rate in HCC patients. CONCLUSIONS The survival model based on the ARGs may be a promising tool to predict the prognosis in HCC patients.
Collapse
Affiliation(s)
- Wanli Yang
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical UniversityXi’an 710032, Shaanxi Province, China
| | - Liaoran Niu
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical UniversityXi’an 710032, Shaanxi Province, China
| | - Xinhui Zhao
- Department of Thyroid and Breast Surgery, Xi’an No. 3 Hospital, The Affiliated Hospital of Northwest University, Northwest UniversityXi’an 710018, Shaanxi Province, China
| | - Lili Duan
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical UniversityXi’an 710032, Shaanxi Province, China
| | - Yiding Li
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical UniversityXi’an 710032, Shaanxi Province, China
| | - Xiaoqian Wang
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical UniversityXi’an 710032, Shaanxi Province, China
| | - Yujie Zhang
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical UniversityXi’an 710032, Shaanxi Province, China
| | - Wei Zhou
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical UniversityXi’an 710032, Shaanxi Province, China
| | - Jinqiang Liu
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical UniversityXi’an 710032, Shaanxi Province, China
| | - Qingchuan Zhao
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical UniversityXi’an 710032, Shaanxi Province, China
| | - Yu Han
- Department of Otolaryngology, Xijing Hospital, Fourth Military Medical UniversityXi’an 710032, Shaanxi Province, China
| | - Daiming Fan
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical UniversityXi’an 710032, Shaanxi Province, China
| | - Liu Hong
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical UniversityXi’an 710032, Shaanxi Province, China
| |
Collapse
|
24
|
Tissue-Specific Impact of Autophagy Genes on the Ubiquitin-Proteasome System in C. elegans. Cells 2020; 9:cells9081858. [PMID: 32784405 PMCID: PMC7464313 DOI: 10.3390/cells9081858] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 07/30/2020] [Accepted: 08/06/2020] [Indexed: 12/30/2022] Open
Abstract
The ubiquitin–proteasome system (UPS) and the autophagy–lysosomal pathway (ALP) are the two main eukaryotic intracellular proteolytic systems involved in maintaining proteostasis. Several studies have reported on the interplay between the UPS and ALP, however it remains largely unknown how compromised autophagy affects UPS function in vivo. Here, we have studied the crosstalk between the UPS and ALP by investigating the tissue-specific effect of autophagy genes on the UPS at an organismal level. Using transgenic Caenorhabditis elegans expressing fluorescent UPS reporters, we show that the downregulation of the autophagy genes lgg-1 and lgg-2 (ATG8/LC3/GABARAP), bec-1 (BECLIN1), atg-7 (ATG7) and epg-5 (mEPG5) by RNAi decreases proteasomal degradation, concomitant with the accumulation of polyubiquitinated proteasomal substrates in a tissue-specific manner. For some of these genes, the changes in proteasomal degradation occur without a detectable alteration in proteasome tissue expression levels. In addition, the lgg-1 RNAi-induced reduction in proteasome activity in intestinal cells is not dependent on sqst-1/p62 accumulation. Our results illustrate that compromised autophagy can affect UPS in a tissue-specific manner, and demonstrate that UPS does not function as a direct compensatory mechanism in an animal. Further, a more profound understanding of the multilayered crosstalk between UPS and ALP can facilitate the development of therapeutic options for various disorders linked to dysfunction in proteostasis.
Collapse
|
25
|
Deng X, Bi Q, Chen S, Chen X, Li S, Zhong Z, Guo W, Li X, Deng Y, Yang Y. Identification of a Five-Autophagy-Related-lncRNA Signature as a Novel Prognostic Biomarker for Hepatocellular Carcinoma. Front Mol Biosci 2020; 7:611626. [PMID: 33505990 PMCID: PMC7831610 DOI: 10.3389/fmolb.2020.611626] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 12/02/2020] [Indexed: 12/21/2022] Open
Abstract
Although great progresses have been made in the diagnosis and treatment of hepatocellular carcinoma (HCC), its prognostic marker remains controversial. In this current study, weighted correlation network analysis and Cox regression analysis showed significant prognostic value of five autophagy-related long non-coding RNAs (AR-lncRNAs) (including TMCC1-AS1, PLBD1-AS1, MKLN1-AS, LINC01063, and CYTOR) for HCC patients from data in The Cancer Genome Atlas. By using them, we constructed a five-AR-lncRNA prognostic signature, which accurately distinguished the high- and low-risk groups of HCC patients. All of the five AR lncRNAs were highly expressed in the high-risk group of HCC patients. This five-AR-lncRNA prognostic signature showed good area under the curve (AUC) value (AUC = 0.751) for the overall survival (OS) prediction in either all HCC patients or HCC patients stratified according to several clinical traits. A prognostic nomogram with this five-AR-lncRNA signature predicted the 3- and 5-year OS outcomes of HCC patients intuitively and accurately (concordance index = 0.745). By parallel comparison, this five-AR-lncRNA signature has better prognosis accuracy than the other three recently published signatures. Furthermore, we discovered the prediction ability of the signature on therapeutic outcomes of HCC patients, including chemotherapy and immunotherapeutic responses. Gene set enrichment analysis and gene mutation analysis revealed that dysregulated cell cycle pathway, purine metabolism, and TP53 mutation may play an important role in determining the OS outcomes of HCC patients in the high-risk group. Collectively, our study suggests a new five-AR-lncRNA prognostic signature for HCC patients.
Collapse
Affiliation(s)
- Xiaoyu Deng
- Institute of Materia Medica, College of Pharmacy, Army Medical University (Third Military Medical University), Chongqing, China
| | - Qinghua Bi
- Institute of Materia Medica, College of Pharmacy, Army Medical University (Third Military Medical University), Chongqing, China
- *Correspondence: Yao Yang
| | - Shihan Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Army Medical University (Third Military Medical University), Chongqing, China
| | - Xianhua Chen
- Diagosis and Treatment Center for Servicemen, The First Affiliated Hospital of Army Medical University (Third Military Medical University), Chongqing, China
| | - Shuhui Li
- Department of Clinical Biochemistry, Faculty of Pharmacy and Laboratory Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhaoyang Zhong
- Cancer Center, Daping Hospital and Research Institute of Surgery, Army Medical University (Third Military Medical University), Chongqing, China
| | - Wei Guo
- Department of Pharmacy, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Xiaohui Li
- Institute of Materia Medica, College of Pharmacy, Army Medical University (Third Military Medical University), Chongqing, China
- Youcai Deng
| | - Youcai Deng
- Institute of Materia Medica, College of Pharmacy, Army Medical University (Third Military Medical University), Chongqing, China
- Xiaohui Li
| | - Yao Yang
- Institute of Materia Medica, College of Pharmacy, Army Medical University (Third Military Medical University), Chongqing, China
- Qinghua Bi
| |
Collapse
|