1
|
Zhang Y, Ding L, Zhang Z, Shen L, Guo Y, Zhang W, Yu Y, Gu Z, Liu J, Kadier A, Geng J, Mao S, Yao X. An Integrated Approach Utilizing Single-Cell and Bulk RNA-Sequencing for the Identification of a Mitophagy-Associated Genes Signature: Implications for Prognostication and Therapeutic Stratification in Prostate Cancer. Biomedicines 2025; 13:311. [PMID: 40002724 PMCID: PMC11853322 DOI: 10.3390/biomedicines13020311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 12/17/2024] [Accepted: 12/24/2024] [Indexed: 02/27/2025] Open
Abstract
Introduction: Prostate cancer, notably prostate adenocarcinoma (PARD), has high incidence and mortality rates. Although typically resistant to immunotherapy, recent studies have found immune targets for prostate cancer. Stratifying patients by molecular subtypes may identify those who could benefit from immunotherapy. Methods: We used single-cell and bulk RNA sequencing data from GEO and TCGA databases. We characterized the tumor microenvironment at the single-cell level, analyzing cell interactions and identifying fibroblasts linked to mitophagy. Target genes were narrowed down at the bulk transcriptome level to construct a PARD prognosis prediction nomogram. Unsupervised consensus clustering classified PARD into subtypes, analyzing differences in clinical features, immune infiltration, and immunotherapy. Furthermore, the cellular functions of the genes of interest were verified in vitro. Results: We identified ten cell types and 160 mitophagy-related single-cell differentially expressed genes (MR-scDEGs). Strong interactions were observed between fibroblasts, endothelial cells, CD8+ T cells, and NK cells. Fibroblasts linked to mitophagy were divided into six subtypes. Intersection of DEGs from three bulk datasets with MR-scDEGs identified 26 key genes clustered into two subgroups. COX regression analysis identified seven prognostic key genes, enabling a prognostic nomogram model. High and low-risk groups showed significant differences in clinical features, immune infiltration, immunotherapy, and drug sensitivity. In prostate cancer cell lines, CAV1, PALLD, and ITGB8 are upregulated, while CLDN7 is downregulated. Knockdown of PALLD significantly inhibits the proliferation and colony-forming ability of PC3 and DU145 cells, suggesting the important roles of this gene in prostate cancer progression. Conclusions: This study analyzed mitophagy-related genes in PARD, predicting prognosis and aiding in subtype identification and immunotherapy response analysis. This approach offers new strategies for treating prostate cancer with specific molecular subtypes and helps develop potential biomarkers for personalized medicine strategies.
Collapse
Affiliation(s)
- Yuke Zhang
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Middle Yan Chang Road, Shanghai 200072, China; (Y.Z.); (L.D.); (Z.Z.); (Y.G.); (W.Z.); (Y.Y.); (Z.G.); (J.L.); (A.K.); (J.G.)
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai 200072, China
| | - Li Ding
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Middle Yan Chang Road, Shanghai 200072, China; (Y.Z.); (L.D.); (Z.Z.); (Y.G.); (W.Z.); (Y.Y.); (Z.G.); (J.L.); (A.K.); (J.G.)
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai 200072, China
| | - Zhijin Zhang
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Middle Yan Chang Road, Shanghai 200072, China; (Y.Z.); (L.D.); (Z.Z.); (Y.G.); (W.Z.); (Y.Y.); (Z.G.); (J.L.); (A.K.); (J.G.)
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai 200072, China
| | - Liliang Shen
- Department of Urology, Ningbo Yinzhou People’s Hospital, 251 Baizhang East Road, Ningbo 315100, China;
| | - Yadong Guo
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Middle Yan Chang Road, Shanghai 200072, China; (Y.Z.); (L.D.); (Z.Z.); (Y.G.); (W.Z.); (Y.Y.); (Z.G.); (J.L.); (A.K.); (J.G.)
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai 200072, China
| | - Wentao Zhang
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Middle Yan Chang Road, Shanghai 200072, China; (Y.Z.); (L.D.); (Z.Z.); (Y.G.); (W.Z.); (Y.Y.); (Z.G.); (J.L.); (A.K.); (J.G.)
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai 200072, China
| | - Yang Yu
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Middle Yan Chang Road, Shanghai 200072, China; (Y.Z.); (L.D.); (Z.Z.); (Y.G.); (W.Z.); (Y.Y.); (Z.G.); (J.L.); (A.K.); (J.G.)
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai 200072, China
| | - Zhuoran Gu
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Middle Yan Chang Road, Shanghai 200072, China; (Y.Z.); (L.D.); (Z.Z.); (Y.G.); (W.Z.); (Y.Y.); (Z.G.); (J.L.); (A.K.); (J.G.)
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai 200072, China
| | - Ji Liu
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Middle Yan Chang Road, Shanghai 200072, China; (Y.Z.); (L.D.); (Z.Z.); (Y.G.); (W.Z.); (Y.Y.); (Z.G.); (J.L.); (A.K.); (J.G.)
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai 200072, China
| | - Aimaitiaji Kadier
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Middle Yan Chang Road, Shanghai 200072, China; (Y.Z.); (L.D.); (Z.Z.); (Y.G.); (W.Z.); (Y.Y.); (Z.G.); (J.L.); (A.K.); (J.G.)
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai 200072, China
| | - Jiang Geng
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Middle Yan Chang Road, Shanghai 200072, China; (Y.Z.); (L.D.); (Z.Z.); (Y.G.); (W.Z.); (Y.Y.); (Z.G.); (J.L.); (A.K.); (J.G.)
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai 200072, China
| | - Shiyu Mao
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Middle Yan Chang Road, Shanghai 200072, China; (Y.Z.); (L.D.); (Z.Z.); (Y.G.); (W.Z.); (Y.Y.); (Z.G.); (J.L.); (A.K.); (J.G.)
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai 200072, China
| | - Xudong Yao
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Middle Yan Chang Road, Shanghai 200072, China; (Y.Z.); (L.D.); (Z.Z.); (Y.G.); (W.Z.); (Y.Y.); (Z.G.); (J.L.); (A.K.); (J.G.)
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai 200072, China
| |
Collapse
|
2
|
Tian S, Song Y, Guo L, Zhao H, Bai M, Miao M. Epigenetic Mechanisms in Osteoporosis: Exploring the Power of m 6A RNA Modification. J Cell Mol Med 2025; 29:e70344. [PMID: 39779466 PMCID: PMC11710941 DOI: 10.1111/jcmm.70344] [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: 09/05/2024] [Revised: 12/12/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Osteoporosis, recognised as a metabolic disorder, has emerged as a significant burden on global health. Although available treatments have made considerable advancements, they remain inadequately addressed. In recent years, the role of epigenetic mechanisms in skeletal disorders has garnered substantial attention, particularly concerning m6A RNA modification. m6A is the most prevalent dynamic and reversible modification in eukaryotes, mediating various metabolic processes of mRNAs, including splicing, structural conversion, translation, translocation and degradation and serves as a crucial component of epigenetic modification. Research has increasingly validated that m6A plays a vital role in the proliferation, differentiation, migration, invasion,and repair of bone marrow mesenchymal stem cells (BMSCs), osteoblasts and osteoclasts, all of which impact the whole process of osteoporosis pathogenesis. Continuous efforts have been made to target m6A regulators and natural products derived from traditional medicine, which exhibit multiple biological activities such as anti-inflammatory and anticancer effects, have emerged as a valuable resources for m6A drug discovery. This paper elaborates on m6A methylation and its regulatory role in osteoporosis, emphasising its implications for diagnosis and treatment, thereby providing theoretical references.
Collapse
Affiliation(s)
- Shuo Tian
- Academy of Traditional Chinese MedicineHenan University of Chinese MedicineZhengzhouChina
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu‐YaoZhengzhouChina
| | - Yagang Song
- Academy of Traditional Chinese MedicineHenan University of Chinese MedicineZhengzhouChina
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu‐YaoZhengzhouChina
| | - Lin Guo
- School of PharmacyHenan University of Chinese MedicineZhengzhouChina
| | - Hui Zhao
- Academy of Traditional Chinese MedicineHenan University of Chinese MedicineZhengzhouChina
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu‐YaoZhengzhouChina
| | - Ming Bai
- Academy of Traditional Chinese MedicineHenan University of Chinese MedicineZhengzhouChina
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu‐YaoZhengzhouChina
| | - Mingsan Miao
- Academy of Traditional Chinese MedicineHenan University of Chinese MedicineZhengzhouChina
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu‐YaoZhengzhouChina
| |
Collapse
|
3
|
Ge Y, Janson V, Liu H. Comprehensive review on leucine-rich pentatricopeptide repeat-containing protein (LRPPRC, PPR protein): A burgeoning target for cancer therapy. Int J Biol Macromol 2024; 282:136820. [PMID: 39476900 DOI: 10.1016/j.ijbiomac.2024.136820] [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: 09/03/2024] [Revised: 10/12/2024] [Accepted: 10/21/2024] [Indexed: 11/04/2024]
Abstract
Leucine-rich pentatricopeptide repeat-containing (LRPPRC), known as the gene mutations that cause Leigh Syndrome French Canadian, encodes a high molecular weight PPR protein (157,905 Da), LRPPRC. LRPPRC binds to DNA, RNA, and proteins to regulate transcription and translation, leading to changes in cell fate. Increasing evidence indicates that LRPPRC plays a pivotal role in various human diseases, particularly cancer in recent years. Here, we review the structure, function, molecular mechanism, as well as inhibitors of LRPPRC. LRPPRC expression elevates in most cancer types and high expression of LRPPRC predicts the poor prognosis of cancer patients. Targeting LRPPRC suppresses tumor progression by affecting several cancer hallmarks, including signal transduction, cancer metabolism, and immune regulation. LRPPRC is a promising target in cancer research, serving as both a biomarker and therapeutic target. Further studies are required to extend the understanding of LRPPRC function and molecular mechanism, as well as to refine novel therapeutic strategies targeting LRPPRC in cancer therapy.
Collapse
Affiliation(s)
- Yunxiao Ge
- Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan 450001, China; China-US (Henan) Hormel Cancer Institute, No.127, Dongming Road, Jinshui District, Zhengzhou, Henan 450008, China
| | - Victor Janson
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Hui Liu
- Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan 450001, China; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; China-US (Henan) Hormel Cancer Institute, No.127, Dongming Road, Jinshui District, Zhengzhou, Henan 450008, China.
| |
Collapse
|
4
|
Yan J, Wang Z, Li Y, Li R, Xiang K. m6A-related genes and their role in Parkinson's disease: Insights from machine learning and consensus clustering. Medicine (Baltimore) 2024; 103:e40484. [PMID: 39533574 PMCID: PMC11557064 DOI: 10.1097/md.0000000000040484] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Parkinson disease (PD) is a chronic neurological disorder primarily characterized by a deficiency of dopamine in the brain. In recent years, numerous studies have highlighted the substantial influence of RNA N6-methyladenosine (m6A) regulators on various biological processes. Nevertheless, the specific contribution of m6A-related genes to the development and progression of PD remains uncertain. In this study, we performed a differential analysis of the GSE8397 dataset in the Gene Expression Omnibus database and selected important m6A-related genes. Candidate m6A-related genes were then screened using a random forest model to predict the risk of PD. A nomogram model was built based on the candidate m6A-related genes. By employing a consensus clustering method, PD was divided into different m6A clusters based on the selected significant m6A-related genes. Finally, we performed immune cell infiltration analysis to explore the immune infiltration between different clusters. We performed a differential analysis of the GSE8397 dataset in the Gene Expression Omnibus database and selected 11 important m6A-related genes. Four candidate m6A-related genes (YTH Domain Containing 2, heterogeneous nuclear ribonucleoprotein C, leucine-rich pentatricopeptide repeat motif containing protein and insulin-like growth factor binding protein-3) were then screened using a random forest model to predict the risk of PD. A nomogram model was built based on the 4 candidate m6A-related genes. The decision curve analysis indicated that patients can benefit from the nomogram model. By employing a consensus clustering method, PD was divided into 2 m6A clusters (cluster A and cluster B) based on the selected significant m6A-related genes. The immune cell infiltration analysis revealed that cluster A and cluster B exhibit distinct immune phenotypes. In conclusion, m6A-related genes play a significant role in the development of PD and our study on m6A clustering may potentially guide personalized treatment strategies for PD in the future.
Collapse
Affiliation(s)
- Jing Yan
- Changchun University of Chinese Medicine, Changchun, Jilin Province, China
| | - Zhengyan Wang
- Changchun University of Chinese Medicine, Changchun, Jilin Province, China
| | - Yunqiang Li
- Changchun University of Chinese Medicine, Changchun, Jilin Province, China
| | - Ruien Li
- Changchun University of Chinese Medicine, Changchun, Jilin Province, China
| | - Ke Xiang
- Jilin Provincial Academy of Chinese Medicine Sciences, Changchun, Jilin Province, China
| |
Collapse
|
5
|
Dushnitzky S, Ishtayeh H, Ashkenazi A. The new kids on the block: RNA-binding proteins regulate autophagy in disease. FEBS J 2024; 291:3811-3819. [PMID: 38825737 DOI: 10.1111/febs.17195] [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: 02/28/2024] [Revised: 04/30/2024] [Accepted: 05/24/2024] [Indexed: 06/04/2024]
Abstract
Mammalian autophagy is a highly regulated and conserved cellular homeostatic process. Its existence allows the degradation of self-components to mediate cell survival in different stress conditions. Autophagy is involved in the regulation of cellular metabolic needs, protecting the cell or tissue from starvation through the degradation and recycling of cytoplasmic materials and organelles to basic molecular building blocks. It also plays a critical role in eliminating damaged or harmful proteins, organelles, and intracellular pathogens. Thus, a deterioration of the process may result in pathological conditions, such as aging-associated disorders and cancer. Understanding the crucial role of autophagy in maintaining the normal physiological function of cells, tissue, or organs has led to copious and expansive research regarding the regulation of this process. So far, most of the research has revolved around transcriptional and post-translational regulation. Here, we discuss the regulation of autophagy-related (ATG) mRNA transcripts by RNA-binding proteins (RBPs). This analysis focuses on how RBPs modulate autophagy in disease. A deeper understanding of the involvement of RBPs in autophagy can facilitate further research and treatment of a variety of human diseases.
Collapse
Affiliation(s)
- Shai Dushnitzky
- The Department of Cell and Developmental Biology, Faculty of Medical & Health Sciences, Tel Aviv University, Israel
| | - Hasan Ishtayeh
- The Department of Cell and Developmental Biology, Faculty of Medical & Health Sciences, Tel Aviv University, Israel
| | - Avraham Ashkenazi
- The Department of Cell and Developmental Biology, Faculty of Medical & Health Sciences, Tel Aviv University, Israel
- Sagol School of Neuroscience, Tel Aviv University, Israel
| |
Collapse
|
6
|
Sun Y, Chen X, Shi Y, Teng F, Dai C, Ge L, Xu J, Jia X. hsa_circ_0020093 suppresses ovarian cancer progression by modulating LRPPRC activity and miR-107/LATS2 signaling. Biol Direct 2024; 19:69. [PMID: 39164777 PMCID: PMC11337591 DOI: 10.1186/s13062-024-00520-y] [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: 05/17/2024] [Accepted: 08/08/2024] [Indexed: 08/22/2024] Open
Abstract
A substantive body of evidence has demonstrated the significant roles of circular RNA (circRNA) in cancer. However, the contribution of dysregulated circRNAs to ovarian cancer (OC) remains elusive. We aim to elucidate the critical roles and mechanisms of hsa_circ_0020093, which was demonstrated to be downregulated in OC tissues in our previous study. In this study, we confirmed the decreased expression of hsa_circ_0020093 in OC tissues and cell lines and demonstrated the negative correlation between its expression and FIGO stage, abdominal implantation and CA125 level of OC patients. Through gain and loss of function studies, we confirmed the inhibitory role of hsa_circ_0020093 in ovarian tumor growth in vitro and in vivo. Mechanistically, based on the peri-nuclear accumulation of hsa_circ_0020093, we discovered the interaction between hsa_circ_0020093 and the mitochondrial protein LRPPRC by RNA pull-down, mass spectrometry, RNA Binding Protein Immunoprecipitation. As a result, qRT-PCR and transmission electron microscopy results showed that the mitochondria mRNA expression and mitochondria abundance were decreased upon hsa_circ_0020093-overexpression. Meanwhile, we also unearthed the hsa_circ_0020093/miR-107/LATS2 axis in OC according to RNA-sequencing, RIP and luciferase reporter assay data. Furthermore, LRPPRC and LATS2 are both reported as the upstream regulators of YAP, our study also studied the crosstalk between hsa_circ_0020093, LRPPRC and miR-107/LATS2, and unearthed the up-regulation of phosphorylated YAP in hsa_circ_0020093-overexpressing OC cells and xenograft tumors. Collectively, our study indicated the novel mechanism of hsa_circ_0020093 in suppressing OC progression through both hsa_circ_0020093/LRPPRC and hsa_circ_0020093/miR-107/LATS2 axes, providing a potential therapeutic target for OC patients.
Collapse
Affiliation(s)
- Yu Sun
- Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), 123 Tianfei Xiang, Mochou Road, Nanjing, 210004, China
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Xiyi Chen
- Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), 123 Tianfei Xiang, Mochou Road, Nanjing, 210004, China
| | - Yaqian Shi
- Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), 123 Tianfei Xiang, Mochou Road, Nanjing, 210004, China
| | - Fang Teng
- Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), 123 Tianfei Xiang, Mochou Road, Nanjing, 210004, China
| | - Chencheng Dai
- Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), 123 Tianfei Xiang, Mochou Road, Nanjing, 210004, China
| | - Lili Ge
- Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), 123 Tianfei Xiang, Mochou Road, Nanjing, 210004, China.
| | - Juan Xu
- Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), 123 Tianfei Xiang, Mochou Road, Nanjing, 210004, China.
| | - Xuemei Jia
- Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), 123 Tianfei Xiang, Mochou Road, Nanjing, 210004, China.
| |
Collapse
|
7
|
Zhang Y, Feng B, Liang Y, Tang Q, Zhang S, Zhang Z, Xu L, Yin J. Prognostic significance of LRPPRC and its association with immune infiltration in liver hepatocellular carcinoma. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL IMMUNOLOGY 2024; 13:105-116. [PMID: 39022790 PMCID: PMC11249856 DOI: 10.62347/xtlj1335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 06/06/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND Leucine rich pentatricopeptide repeat containing (LRPPRC) protein is a multifunctional protein involved in cell cycle progression and tumor development. However, its prognostic significance and association with immune infiltration in Liver hepatocellular carcinoma (LIHC) remain unclear. METHODS We utilized transcriptomic and clinical data from The Cancer Genome Atlas (TCGA) and Genotype Tissue Expression (GTEx) databases of LIHC patients to investigate the potential pro-cancer role of LRPPRC, including differential expression of LRPPRC in LIHC, prognostic value, clinicopathological features, immune cell infiltration relevance and function enrichment analysis. RESULTS Our findings suggest that LRPPRC is upregulated in LIHC and exhibits correlations with survival, clinical stage, and tumor grade in LIHC patients. Additionally, immune infiltration analysis revealed significant negative correlations between LRPPRC expression and multiple tumor-infiltrating immune cells, including CTLs, DCs, pDCs, B cells, Th17 cells, neutrophils, T cells, Mast cells, Th1 cells, Tregs, and NK cells, whereas a significant positive correlation was observed with infiltration of Th2 cells, T helper cells and Tcms. Furthermore, functional enrichment analysis indicated that LRPPRC may be involved in G2m checkpoint, mitotic spindle, E2f targets, Wnt Beta catenin signaling, spermatogenesis and other processes.
Collapse
Affiliation(s)
- Yanqiu Zhang
- Center for Clinical Laboratory, The First Affiliated Hospital of Soochow UniversitySuzhou, Jiangsu, The People’s Republic of China
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune MedicineHefei, Anhui, The People’s Republic of China
| | - Bin Feng
- Center for Clinical Laboratory, The First Affiliated Hospital of Soochow UniversitySuzhou, Jiangsu, The People’s Republic of China
| | - Yuting Liang
- Center for Clinical Laboratory, The First Affiliated Hospital of Soochow UniversitySuzhou, Jiangsu, The People’s Republic of China
| | - Qingqin Tang
- Center for Clinical Laboratory, The First Affiliated Hospital of Soochow UniversitySuzhou, Jiangsu, The People’s Republic of China
| | - Sheng Zhang
- Center for Clinical Laboratory, The First Affiliated Hospital of Soochow UniversitySuzhou, Jiangsu, The People’s Republic of China
| | - Zheng Zhang
- Center for Clinical Laboratory, The First Affiliated Hospital of Soochow UniversitySuzhou, Jiangsu, The People’s Republic of China
| | - Li Xu
- Department of Pharmacy, The First Affiliated Hospital of Soochow UniversitySuzhou, Jiangsu, The People’s Republic of China
| | - Jingping Yin
- Center for Clinical Laboratory, The First Affiliated Hospital of Soochow UniversitySuzhou, Jiangsu, The People’s Republic of China
| |
Collapse
|
8
|
Zhao Z, Sun Y, Tang J, Yang Y, Xu X. LRPPRC regulates malignant behaviors, protects mitochondrial homeostasis, mitochondrial function in osteosarcoma and derived cancer stem-like cells. BMC Cancer 2023; 23:935. [PMID: 37789316 PMCID: PMC10548780 DOI: 10.1186/s12885-023-11443-8] [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: 12/24/2022] [Accepted: 09/25/2023] [Indexed: 10/05/2023] Open
Abstract
BACKGROUND Leucine-rich pentatricopeptide repeat containing (LRPPRC) is a potential oncogene in multiple tumor types, including lung adenocarcinoma, esophageal squamous cell carcinoma and gastric cancer. LRPPRC exerts its tumor-promoting effects mainly by regulating mitochondrial homeostasis and inducing oxidative stress. However, the exact role and mechanisms by which LRPPRC acts in osteosarcoma and osteosarcoma-derived cancer stem-like cells (CSCs), which potentially critically contribute to recurrence, metastasis and chemoresistance, are still largely unclear. METHODS LRPPRC level in osteosarcoma cells and CSCs were detected by western blot. Effects of LRPPRC on CSCs were accessed after LRPPRC knockdown by introducing lentivirus containing shRNA targeting to LRPPRC mRNA. RESULTS we found that LRPPRC was highly expressed in several osteosarcoma cell lines and that LRPPRC knockdown inhibited malignant behaviors, including proliferation, invasion, colony formation and tumor formation, in MG63 and U2OS cells. Enriched CSCs derived from MG63 and U2OS cells presented upregulated LRPPRC levels compared to parental cells (PCs), and LRPPRC knockdown markedly decreased the sphere-forming capacity. These findings demonstrate that LRPPRC knockdown decreased stemness in CSCs. Consistent with a previous report, LRPPRC knockdown decreased the expression levels of FOXM1 and its downstream target genes, including PRDX3, MnSOD and catalase, which are responsible for scavenging reactive oxygen species (ROS). Expectedly, LRPPRC knockdown increased the accumulation of ROS in osteosarcoma and osteosarcoma-derived CSCs under hypoxic conditions due to the decrease in ROS scavenging proteins. Moreover, LRPPRC knockdown sensitized osteosarcomas and CSCs against carboplatin, a ROS-inducing chemoagent, and promoted apoptosis. Furthermore, LRPPRC knockdown significantly decreased the mitochondrial membrane potential, disturbed mitochondrial homeostasis and led to mitochondrial dysfunction. CONCLUSION Taken together, these findings indicated that LRPPRC exerts critical roles in regulating mitochondrial homeostasis, mitochondrial function and tumorigenesis in osteosarcomas and osteosarcoma-derived CSCs. This suggests that LRPPRC might be a promising therapeutic target for osteosarcomas.
Collapse
Affiliation(s)
- Ziyi Zhao
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610041, P.R. China
| | - Yingwei Sun
- Department of Pharmacy, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610041, P.R. China
| | - Jing Tang
- Chongqing Three gorges medical college, Chongqing, 404120, P.R. China
| | - Yuting Yang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610041, P.R. China
| | - Xiaochao Xu
- College of Food and Biological Engineering, Chengdu University, Chengdu, 610000, P.R. China.
| |
Collapse
|
9
|
Liang L, Sun W, Wei X, Wang L, Ruan H, Zhang J, Li S, Zhao B, Li M, Cai Z, Huang J. Oxymatrine suppresses colorectal cancer progression by inhibiting NLRP3 inflammasome activation through mitophagy induction in vitro and in vivo. Phytother Res 2023; 37:3342-3362. [PMID: 36974424 DOI: 10.1002/ptr.7808] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/07/2023] [Accepted: 03/07/2023] [Indexed: 03/29/2023]
Abstract
Chinese herb Radix sophorae tonkinensis extract oxymatrine shows anticancer effects. This study evaluated the role of oxymatrine in colorectal cancer (CRC) and the underlying molecular events in vitro and in vivo. CRC cells were treated with different doses of oxymatrine to assess cell viability, reactive oxygen species production, gene expression, and gene alterations. Meanwhile, mouse xenograft and liver metastasis models were used to assess the effects of oxymatrine using histology examination, transmission electron microscopy, and Western blot, respectively. Our results showed that oxymatrine treatment triggered CRC cell mitophagy to inhibit CRC cell growth, migration, invasion, and metastasis in vitro and in vivo. At the gene level, oxymatrine inhibited LRPPRC to promote Parkin translocation into the mitochondria and reduce the mitophagy-activated NLRP3 inflammasome. Thus, oxymatrine had an anticancer activity through LRPPRC inhibition, mitophagy induction, and NLRP3 inflammasome suppression in the CRC cell xenograft and liver metastasis models. In conclusion, the study demonstrates the oxymatrine anti- CRC activity through its unique role in regulating CRC cell mitophagy and NLRP3 inflammasome levels in vitro and in vivo.
Collapse
Affiliation(s)
- Li Liang
- Department of Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Weiliang Sun
- Department of Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiaoxuan Wei
- Department of Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Li Wang
- Department of Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Huaqiang Ruan
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Junchuan Zhang
- Department of Traditional Chinese Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Suyan Li
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Bi Zhao
- Department of Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Mengshi Li
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhengwen Cai
- Department of Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jie'an Huang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
10
|
Xu Z, Li X, Ding Z, Zhang Y, Peng Z, Yang X, Cao W, Du R. LRPPRC inhibits autophagy and promotes foam cell formation in atherosclerosis. FEBS J 2022; 289:7545-7560. [PMID: 35792704 DOI: 10.1111/febs.16567] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 05/13/2022] [Accepted: 06/27/2022] [Indexed: 01/14/2023]
Abstract
Lipid-laden macrophages are considered as the main source of foam cells in atherosclerosis; however, the mechanism for macrophage foam cell formation remains unknown. Here, we explore the mechanism behind foam cell formation to potentially identify a novel treatment for atherosclerosis. Our data demonstrated that leucine-rich pentatricopeptide repeat-containing protein (LRPPRC) increased in the atherosclerotic plaques of LDLR-/- mice fed with a Western diet. LRPPRC was also upregulated in mice peritoneal macrophages and RAW 264.7 cells treated with oxidative low density lipoprotein, whereas knockdown of LRPPRC by transfecting with small interfering (Si)-LRPPRC in RAW 264.7 cells decreased foam cell formation. Furthermore, Si-LRPPRC promoted autophagy and increased the expression of cholesterol efflux protein ATP-binding cassette transporter A1 in RAW 264.7 cells. Moreover, intervention with MHY1485 in RAW 264.7 cells revealed that autophagy was inhibited by LRPPRC via the Akt-mechanistic target of rapamycin pathway. Taken together, we confirm for the first time that LRPPRC is increased within the atherosclerotic plaques of mice and enhances the process of foam cell formation. The knockdown of LRPPRC inhibited foam cell formation by activating macrophage autophagy. Our findings indicate that the regulation of macrophage LRPPRC expression may be a novel strategy for ameliorating atherosclerosis.
Collapse
Affiliation(s)
- Zhou Xu
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Xinran Li
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Zhiquan Ding
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Yuyang Zhang
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Zhiwei Peng
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Xin Yang
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Wangsen Cao
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Ronghui Du
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China.,State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| |
Collapse
|
11
|
Yang Y, Yuan H, Zhao L, Guo S, Hu S, Tian M, Nie Y, Yu J, Zhou C, Niu J, Wang G, Song Y. Targeting the miR-34a/LRPPRC/MDR1 axis collapse the chemoresistance in P53 inactive colorectal cancer. Cell Death Differ 2022; 29:2177-2189. [PMID: 35484333 PMCID: PMC9613927 DOI: 10.1038/s41418-022-01007-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 04/11/2022] [Accepted: 04/11/2022] [Indexed: 11/09/2022] Open
Abstract
P53 mutation is an important cause of chemoresistance in colorectal cancer (CRC). The investigation and identification of the downstream targets and underlying molecular mechanism of chemoresistance induced by P53 abnormalities are therefore of great clinical significance. In this study, we demonstrated and reported for the first time that leucine-rich pentatricopeptide repeat-containing protein (LRPPRC) is a key functional downstream factor and therapeutic target for P53 mutation-induced chemoresistance. Due to its RNA binding function, LRPPRC specifically bound to the mRNA of multidrug resistance 1 (MDR1), increasing MDR1 mRNA stability and protein expression. In normal cells, P53 induced by chemotherapy inhibited the expression of LRPPRC via miR-34a and in turn reduced the expression of MDR1. However, chemotherapy-induced P53/miR-34a/LRPPRC/MDR1 signalling pathway activation was lost when P53 was mutated. Additionally, the accumulated LRPPRC and MDR1 promoted drug resistance. Most importantly, gossypol-acetic acid (GAA) was recently reported by our team as the first specific inhibitor of LRPPRC. In CRC cells with P53 mutation, GAA effectively induced degradation of the LRPPRC protein and reduced chemoresistance. Both in vivo and in vitro experiments revealed that combination chemotherapy with GAA and 5-fluorouracil (5FU) yielded improved treatment outcomes. In this study, we reported a novel mechanism and target related to P53-induced drug resistance and provided corresponding interventional strategies for the precision treatment of CRC.
Collapse
Affiliation(s)
- Yang Yang
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
- Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050051, China
| | - Hongyu Yuan
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lianmei Zhao
- Research center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Shichao Guo
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Sijun Hu
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710000, China
| | - Miaomiao Tian
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710000, China
| | - Yongzhan Nie
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710000, China
| | - Jiarui Yu
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chaoxi Zhou
- The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
| | - Jian Niu
- The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
| | - Guiying Wang
- The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China.
- Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050051, China.
| | - Yongmei Song
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
12
|
Gayan S, Joshi G, Dey T. Biomarkers of mitochondrial origin: a futuristic cancer diagnostic. Integr Biol (Camb) 2022; 14:77-88. [PMID: 35780307 DOI: 10.1093/intbio/zyac008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 05/17/2022] [Accepted: 05/27/2022] [Indexed: 11/12/2022]
Abstract
Cancer is a highly fatal disease without effective early-stage diagnosis and proper treatment. Along with the oncoproteins and oncometabolites, several organelles from cancerous cells are also emerging as potential biomarkers. Mitochondria isolated from cancer cells are one such biomarker candidates. Cancerous mitochondria exhibit different profiles compared with normal ones in morphology, genomic, transcriptomic, proteomic and metabolic landscape. Here, the possibilities of exploring such characteristics as potential biomarkers through single-cell omics and Artificial Intelligence (AI) are discussed. Furthermore, the prospects of exploiting the biomarker-based diagnosis and its futuristic utilization through circulatory tumor cell technology are analyzed. A successful alliance of circulatory tumor cell isolation protocols and a single-cell omics platform can emerge as a next-generation diagnosis and personalized treatment procedure.
Collapse
Affiliation(s)
- Sukanya Gayan
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, India
| | - Gargee Joshi
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, India
| | - Tuli Dey
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, India
| |
Collapse
|
13
|
Ashrafizadeh M, Paskeh MDA, Mirzaei S, Gholami MH, Zarrabi A, Hashemi F, Hushmandi K, Hashemi M, Nabavi N, Crea F, Ren J, Klionsky DJ, Kumar AP, Wang Y. Targeting autophagy in prostate cancer: preclinical and clinical evidence for therapeutic response. J Exp Clin Cancer Res 2022; 41:105. [PMID: 35317831 PMCID: PMC8939209 DOI: 10.1186/s13046-022-02293-6] [Citation(s) in RCA: 96] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 02/16/2022] [Indexed: 02/08/2023] Open
Abstract
Prostate cancer is a leading cause of death worldwide and new estimates revealed prostate cancer as the leading cause of death in men in 2021. Therefore, new strategies are pertinent in the treatment of this malignant disease. Macroautophagy/autophagy is a “self-degradation” mechanism capable of facilitating the turnover of long-lived and toxic macromolecules and organelles. Recently, attention has been drawn towards the role of autophagy in cancer and how its modulation provides effective cancer therapy. In the present review, we provide a mechanistic discussion of autophagy in prostate cancer. Autophagy can promote/inhibit proliferation and survival of prostate cancer cells. Besides, metastasis of prostate cancer cells is affected (via induction and inhibition) by autophagy. Autophagy can affect the response of prostate cancer cells to therapy such as chemotherapy and radiotherapy, given the close association between autophagy and apoptosis. Increasing evidence has demonstrated that upstream mediators such as AMPK, non-coding RNAs, KLF5, MTOR and others regulate autophagy in prostate cancer. Anti-tumor compounds, for instance phytochemicals, dually inhibit or induce autophagy in prostate cancer therapy. For improving prostate cancer therapy, nanotherapeutics such as chitosan nanoparticles have been developed. With respect to the context-dependent role of autophagy in prostate cancer, genetic tools such as siRNA and CRISPR-Cas9 can be utilized for targeting autophagic genes. Finally, these findings can be translated into preclinical and clinical studies to improve survival and prognosis of prostate cancer patients. • Prostate cancer is among the leading causes of death in men where targeting autophagy is of importance in treatment; • Autophagy governs proliferation and metastasis capacity of prostate cancer cells; • Autophagy modulation is of interest in improving the therapeutic response of prostate cancer cells; • Molecular pathways, especially involving non-coding RNAs, regulate autophagy in prostate cancer; • Autophagy possesses both diagnostic and prognostic roles in prostate cancer, with promises for clinical application.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, 34956, Istanbul, Turkey.
| | - Mahshid Deldar Abad Paskeh
- 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
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | | | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, 34396, Istanbul, Turkey
| | - Farid Hashemi
- Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, 1417466191, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine University of Tehran, Tehran, Iran
| | - 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
| | - Noushin Nabavi
- Department of Urological Sciences and Vancouver Prostate Centre, University of British Columbia, V6H3Z6, Vancouver, BC, Canada
| | - Francesco Crea
- Cancer Research Group-School of Life Health and Chemical Sciences, The Open University, Walton Hall, Milton Keynes, MK7 6AA, UK
| | - Jun Ren
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA.,Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Daniel J Klionsky
- Life Sciences Institute & Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Alan Prem Kumar
- Cancer Science Institute of Singapore and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore. .,NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Yuzhuo Wang
- Department of Urological Sciences and Vancouver Prostate Centre, University of British Columbia, V6H3Z6, Vancouver, BC, Canada.
| |
Collapse
|
14
|
Bchetnia M, Tardif J, Morin C, Laprise C. Expression signature of the Leigh syndrome French-Canadian type. Mol Genet Metab Rep 2022; 30:100847. [PMID: 35242578 PMCID: PMC8856909 DOI: 10.1016/j.ymgmr.2022.100847] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 01/29/2022] [Indexed: 10/28/2022] Open
|
15
|
Wei WS, Wang N, Deng MH, Dong P, Liu JY, Xiang Z, Li XD, Li ZY, Liu ZH, Peng YL, Li Z, Jiang LJ, Yao K, Ye YL, Lu WH, Zhang ZL, Zhou FJ, Liu ZW, Xie D, Yu CP. LRPPRC regulates redox homeostasis via the circANKHD1/FOXM1 axis to enhance bladder urothelial carcinoma tumorigenesis. Redox Biol 2021; 48:102201. [PMID: 34864630 PMCID: PMC8645923 DOI: 10.1016/j.redox.2021.102201] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/25/2021] [Accepted: 11/26/2021] [Indexed: 01/03/2023] Open
Abstract
Reactive oxygen species (ROS) which are continuously generated mainly by mitochondria, have been proved to play an important role in the stress signaling of cancer cells. Moreover, pentatricopeptide repeat (PPR) proteins have been suggested to take part in mitochondrial metabolism. However, the mechanisms integrating the actions of these distinct networks in urothelial carcinoma of the bladder (UCB) pathogenesis are elusive. In this study, we found that leucine rich pentatricopeptide repeat containing (LRPPRC) was frequently upregulated in UCB and that it was an independent prognostic factor in UCB. We further revealed that LRPPRC promoted UCB tumorigenesis by regulating the intracellular ROS homeostasis. Mechanistically, LRPPRC modulates ROS balance and protects UCB cells from oxidative stress via mt-mRNA metabolism and the circANKHD1/FOXM1 axis. In addition, the SRA stem-loop interacting RNA binding protein (SLIRP) directly interacted with LRPPRC to protect it from ubiquitination and proteasomal degradation. Notably, we showed that LRPPRC modulated the tumorigenesis of UCB cells in a circANKHD1-FOXM1-dependent manner. In conclusion, LRPPRC exerts critical roles in regulating UCB redox homeostasis and tumorigenesis, and is a prognostic factor for UCB; suggesting that LRPPRC may serve as an exploitable therapeutic target in UCB.
Collapse
Affiliation(s)
- Wen-Su Wei
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, PR China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, PR China
| | - Ning Wang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, PR China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, PR China
| | - Min-Hua Deng
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, PR China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, PR China
| | - Pei Dong
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, PR China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, PR China
| | - Jian-Ye Liu
- Department of Urology, Xiangya Third Hospital, No. 106, 2nd Zhongshan Road, Changsha, PR China
| | - Zhen Xiang
- Fudan University Shanghai Cancer Center, Shanghai, 200032, PR China
| | - Xiang-Dong Li
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, PR China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, PR China
| | - Zhi-Yong Li
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, PR China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, PR China
| | - Zhen-Hua Liu
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, PR China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, PR China
| | - Yu-Lu Peng
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, PR China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, PR China
| | - Zhen Li
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, PR China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, PR China
| | - Li-Juan Jiang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, PR China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, PR China
| | - Kai Yao
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, PR China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, PR China
| | - Yun-Lin Ye
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, PR China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, PR China
| | - Wen-Hua Lu
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, PR China
| | - Zhi-Ling Zhang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, PR China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, PR China
| | - Fang-Jian Zhou
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, PR China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, PR China
| | - Zhuo-Wei Liu
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, PR China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, PR China.
| | - Dan Xie
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, PR China; Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, PR China.
| | - Chun-Ping Yu
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, PR China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, PR China.
| |
Collapse
|
16
|
m6A-Mediated Tumor Invasion and Methylation Modification in Breast Cancer Microenvironment. JOURNAL OF ONCOLOGY 2021; 2021:9987376. [PMID: 34745261 PMCID: PMC8566073 DOI: 10.1155/2021/9987376] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/17/2021] [Accepted: 09/20/2021] [Indexed: 12/11/2022]
Abstract
Background We analyzed the n6-methyladenosine (m6A) modification patterns of immune cells infiltrating the tumor microenvironment of breast cancer (BC) to provide a new perspective for the early diagnosis and treatment of BC. Methods Based on 23 m6A regulatory factors, we identified m6A-related gene characteristics and m6A modification patterns in BC through unsupervised cluster analysis. To examine the differences in biological processes among various m6A modification modes, we performed genomic variation analysis. We then quantified the relative infiltration levels of different immune cell subpopulations in the tumor microenvironment of BC using the CIBERSORT algorithm and single-sample gene set enrichment analysis. Univariate Cox analysis was used to screen for m6A characteristic genes related to prognosis. Finally, we evaluated the m6A modification pattern of patients with a single BC by constructing the m6Ascore based on principal component analysis. Results We identified three different m6A modification patterns in 2128 BC samples. A higher abundance of the immune infiltration of the m6Acluster C was indicated by the results of CIBERSORT and the single-sample gene set enrichment analysis. Based on the m6A characteristic genes obtained through screening, the m6Ascore was determined. The BC patients were segregated into m6Ascore groups of low and high categories, which revealed significant survival benefits among patients with low m6Ascores. Additionally, the high-m6Ascore group had a higher mutation frequency and was associated with low PD-L1 expression, and the m6Ascore and tumor mutation burden showed a positive correlation. In addition, treatment effects were better in patients in the high-m6Ascore group. Conclusions In case of a single patient with BC, the immune cell infiltration characteristics of the tumor microenvironment and the m6A methylation modification pattern could be evaluated using the m6Ascore. Our results provide a foundation for improving personalized immunotherapy of BC.
Collapse
|
17
|
Zhao H, Zhou Q, Shi C, Shao Y, Ni J, Lou J, Wei S. RNA N6-Methyladenosine Patterns in Hepatocellular Carcinoma Reveal a Distinct Immune Infiltration Landscape and Clinical Significance. Med Sci Monit 2021; 27:e930994. [PMID: 34690344 PMCID: PMC8555444 DOI: 10.12659/msm.930994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND RNA N6-methyladenosine (m6A) methylation, the most abundant and prominent form of epigenetic modification, is involved in hepatocellular carcinoma (HCC) initiation and progression. However, the role of m6A methylation in HCC tumor microenvironment (TME) formation is unexplored. This study aimed to reveal the TME features of HCC patients with distinct m⁶A expression patterns and establish a prognostic model based on m⁶A signatures for HCC cohorts. MATERIAL AND METHODS We classified the m⁶A methylation patterns in 365 HCC samples based on 21 m6A modulators using a consensus clustering algorithm. Single-sample gene set enrichment analysis algorithm was used to quantify the abundance of immune cell infiltration. Gene set variation analysis revealed the biological characteristics between the m⁶A modification patterns. The m6A-based prognostic model was constructed using a training set with least absolute shrinkage and selection operator regression and validated in internal and external datasets. RESULTS Two distinct m⁶A modification patterns exhibiting different TME immune-infiltrating characteristics, heterogeneity, and prognostic variations were identified in the HCC cohort. After depicting the immune landscape of TME in HCC, we found patients with high LRPPRC m⁶A modulator expression had depletion of T cells, cytotoxic cells, dendritic cells, and cytolytic activity response. A high m⁶A score, characterized by suppression of immunity, indicated an immune-excluded TME phenotype, with poor survival. A nomogram was developed to facilitate HCC clinical decision making. CONCLUSIONS Our results highlight the nonnegligible role of m6A methylation in TME formation and reveal a potential clinical application of the m⁶A-associated prognostic model for patients with HCC.
Collapse
Affiliation(s)
- Hua Zhao
- Department of Geriatrics, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China (mainland)
| | - Qiujun Zhou
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China (mainland)
| | - Chengwei Shi
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China (mainland)
| | - Yaojian Shao
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China (mainland)
| | - Junjie Ni
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China (mainland)
| | - Jianying Lou
- Department of Hepato-Pancreato-Biliary Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China (mainland)
| | - Shenyu Wei
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China (mainland)
| |
Collapse
|
18
|
Esparza-Moltó PB, Cuezva JM. Reprogramming Oxidative Phosphorylation in Cancer: A Role for RNA-Binding Proteins. Antioxid Redox Signal 2020; 33:927-945. [PMID: 31910046 DOI: 10.1089/ars.2019.7988] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Significance: Cancer is a major disease imposing high personal and economic burden draining large part of National Health Care and Research budgets worldwide. In the last decade, research in cancer has underscored the reprogramming of metabolism to an enhanced aerobic glycolysis as a major trait of the cancer phenotype with great potential for targeted therapy. Recent Advances: Mitochondria are essential organelles in metabolic reprogramming for controlling the production of biological energy through oxidative phosphorylation (OXPHOS) and the supply of metabolic precursors that sustain proliferation. In addition, mitochondria are critical hubs that integrate different signaling pathways that control cellular metabolism and cell fate. The mitochondrial ATP synthase plays a fundamental role in OXPHOS and cellular signaling. Critical Issues: This review overviews mitochondrial metabolism and OXPHOS, and the major changes reported in the expression and function of mitochondrial proteins of OXPHOS in oncogenesis and in cellular differentiation. We summarize the prominent role that RNA-binding proteins (RNABPs) play in the sorting and localized translation of nuclear-encoded mRNAs that help define the mitochondrial cell-type-specific phenotype. Moreover, we emphasize the mechanisms that contribute to restrain the activity and expression of the mitochondrial ATP synthase in carcinomas, and illustrate that the dysregulation of proteins that control energy metabolism correlates with patients' survival. Future Directions: Future research should elucidate the mechanisms and RNABPs that promote the specific alterations of the mitochondrial phenotype in carcinomas arising from different tissues with the final aim of developing new therapeutic strategies to treat cancer.
Collapse
Affiliation(s)
- Pau B Esparza-Moltó
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, Madrid, Spain
| | - José M Cuezva
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
19
|
Li W, Dai Y, Shi B, Yue F, Zou J, Xu G, Jiang X, Wang F, Zhou X, Liu L. LRPPRC sustains Yap-P27-mediated cell ploidy and P62-HDAC6-mediated autophagy maturation and suppresses genome instability and hepatocellular carcinomas. Oncogene 2020; 39:3879-3892. [PMID: 32203162 DOI: 10.1038/s41388-020-1257-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 02/28/2020] [Accepted: 03/03/2020] [Indexed: 12/25/2022]
Abstract
Mutants in the gene encoding mitochondrion-associated protein LRPPRC were found to be associated with French Canadian Type Leigh syndrome, a human disorder characterized with neurodegeneration and cytochrome c oxidase deficiency. LRPPRC interacts with one of microtubule-associated protein family MAP1S that promotes autophagy initiation and maturation to suppress genomic instability and tumorigenesis. Previously, although various studies have attributed LRPPRC nuclear acid-associated functions, we characterized that LRPPRC acted as an inhibitor of autophagy in human cancer cells. Here we show that liver-specific deletion of LRPPRC causes liver-specific increases of YAP and P27 and decreases of P62, leading to an increase of cell polyploidy and an impairment of autophagy maturation. The blockade of autophagy maturation and promotion of polyploidy caused by LRPPRC depletion synergistically enhances diethylnitrosamine-induced DNA damage, genome instability, and further tumorigenesis so that LRPPRC knockout mice develop more and larger hepatocellular carcinomas and survive a shorter lifespan. Therefore, LRPPRC suppresses genome instability and hepatocellular carcinomas and promotes survivals in mice by sustaining Yap-P27-mediated cell ploidy and P62-HDAC6-controlled autophagy maturation.
Collapse
Affiliation(s)
- Wenjiao Li
- The Fifth Affiliated Hospital of Guangzhou Medical University, 621 Gangwan Rd, Huangpu District, Guangzhou, 710700, Guangdong, PR China
- Institute of Biosciences and Technology, Texas A&M University, 2121 W. Holcombe Blvd., Houston, TX, 77030, USA
| | - Yuan Dai
- Institute of Biosciences and Technology, Texas A&M University, 2121 W. Holcombe Blvd., Houston, TX, 77030, USA
| | - Boyun Shi
- The Fifth Affiliated Hospital of Guangzhou Medical University, 621 Gangwan Rd, Huangpu District, Guangzhou, 710700, Guangdong, PR China
- Institute of Biosciences and Technology, Texas A&M University, 2121 W. Holcombe Blvd., Houston, TX, 77030, USA
| | - Fei Yue
- Institute of Biosciences and Technology, Texas A&M University, 2121 W. Holcombe Blvd., Houston, TX, 77030, USA
| | - Jing Zou
- Institute of Biosciences and Technology, Texas A&M University, 2121 W. Holcombe Blvd., Houston, TX, 77030, USA
| | - Guibin Xu
- The Fifth Affiliated Hospital of Guangzhou Medical University, 621 Gangwan Rd, Huangpu District, Guangzhou, 710700, Guangdong, PR China
- Institute of Biosciences and Technology, Texas A&M University, 2121 W. Holcombe Blvd., Houston, TX, 77030, USA
| | - Xianhan Jiang
- The Fifth Affiliated Hospital of Guangzhou Medical University, 621 Gangwan Rd, Huangpu District, Guangzhou, 710700, Guangdong, PR China
- Institute of Biosciences and Technology, Texas A&M University, 2121 W. Holcombe Blvd., Houston, TX, 77030, USA
| | - Fen Wang
- Institute of Biosciences and Technology, Texas A&M University, 2121 W. Holcombe Blvd., Houston, TX, 77030, USA
| | - Xinke Zhou
- The Fifth Affiliated Hospital of Guangzhou Medical University, 621 Gangwan Rd, Huangpu District, Guangzhou, 710700, Guangdong, PR China
| | - Leyuan Liu
- The Fifth Affiliated Hospital of Guangzhou Medical University, 621 Gangwan Rd, Huangpu District, Guangzhou, 710700, Guangdong, PR China.
- Institute of Biosciences and Technology, Texas A&M University, 2121 W. Holcombe Blvd., Houston, TX, 77030, USA.
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
20
|
Jiang G, Liang X, Huang Y, Lan Z, Zhang Z, Su Z, Fang Z, Lai Y, Yao W, Liu T, Hu L, Wang F, Huang H, Liu L, Jiang X. p62 promotes proliferation, apoptosis‑resistance and invasion of prostate cancer cells through the Keap1/Nrf2/ARE axis. Oncol Rep 2020; 43:1547-1557. [PMID: 32323805 PMCID: PMC7107779 DOI: 10.3892/or.2020.7527] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 01/17/2020] [Indexed: 12/19/2022] Open
Abstract
Prostate cancer poses a public health threat to hundreds of people around the world. p62 has been identified as a tumor suppressor, however, the mechanism by which p62 promotes prostate cancer remains poorly understood. The present study aimed to investigate whether p62 promotes proliferation, apoptosis resistance and invasion of prostate cancer cells via the Kelch-like ECH-associated protein 1/nuclear factor erytheroid-derived 2-like 2/antioxidant response element (Keap1/Nrf2/ARE) axis. Immunohistochemical staining and immunoblotting were performed to determine the protein levels. Rates of proliferation, invasion and apoptosis of prostate cancer cells were assessed using an RTCA system and flow cytometric assays. Levels of reactive oxygen species (ROS) were assessed using Cell ROX Orange reagent and mRNA levels of Nrf2 target genes were detected by qRT-PCR. It was revealed that p62 increased the levels and activities of Nrf2 by suppressing Keap1-mediated proteasomal degradation in prostate cancer cells and tissues, and high levels of p62 promoted growth of prostate cancer through the Keap1/Nrf2/ARE system. Silencing of Nrf2 in DU145 cells overexpressing p62 led to decreases in the rate of cell proliferation and invasion and an increase in the rate of cell apoptosis. p62 activated the Nrf2 pathway, promoted the transcription of Nrf2-mediated target genes and suppressed ROS in prostate cancer. Therefore, p62 promoted the development of prostate cancer by activating the Keap1/Nrf2/ARE pathway and decreasing p62 may provide a new strategy to ameliorate tumor aggressiveness and suppress tumorigenesis to improve clinical outcomes.
Collapse
Affiliation(s)
- Ganggang Jiang
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| | - Xue Liang
- Department of Center Laboratory, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| | - Yiqiao Huang
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| | - Ziquan Lan
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| | - Zhiming Zhang
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| | - Zhengming Su
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| | - Zhiyuan Fang
- Department of Center Laboratory, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| | - Yuxiong Lai
- Department of Center Laboratory, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| | - Wenxia Yao
- Department of Center Laboratory, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| | - Ting Liu
- Department of Center Laboratory, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| | - La Hu
- Department of Center Laboratory, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| | - Fen Wang
- Center for Translational Cancer Research, Texas A&M Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030, USA
| | - Hai Huang
- Department of Urology, The Sun Yat‑sen Memorial Hospital, Sun Yat‑sen University, Guangzhou 510120, P.R. China
| | - Leyuan Liu
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| | - Xianhan Jiang
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| |
Collapse
|
21
|
Yu K, Xiang L, Li S, Wang S, Chen C, Mu H. HIF1α promotes prostate cancer progression by increasing ATG5 expression. Anim Cells Syst (Seoul) 2019; 23:326-334. [PMID: 31700698 PMCID: PMC6830197 DOI: 10.1080/19768354.2019.1658637] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 06/09/2019] [Accepted: 07/26/2019] [Indexed: 01/07/2023] Open
Abstract
Prostate cancer (PCa) is the most frequently diagnosed cancer among men. However, the major modifiable risk factors for PCa are poorly known and its specific mechanism of progression remains unclear. Here we reported that, in prostate cancer cells, the autophagy level was elevated under hypoxic condition, as well as the mRNA and protein level of ATG5, which is an important gene related to autophagy. Furthermore, we found HIF1α could directly bind to the promoter of ATG5 and promote the expression of ATG5 on transcriptional level by luciferase assay and ChIP assay. Intriguingly, overexpression of HIF1α by HIF1α-M could increase tumor size and the effect could be abolished by knockdown ATG5 by si-ATG5 in BALB/cA-nu/nu nude mice. Importantly, HIF1α could also promote the metastasis of PC-3 cells by upregulating the ATG5 and autophagy level and knockdown ATG5 and inhibition autophagy both could abolish the effect of overexpression of HIF1α on the migration of PC-3 cells. Taken together, our results, for the first time, proved that HIF1α could promote the proliferation and migration of PC-3 cells by direct upregulating ATG5 and autophagy level in PC-3 prostate cancer cells. Our findings not only provide new perspective for the relationship between hypoxia and autophagy, but also add new potential therapeutic regimens for the treatment of prostate cancers.
Collapse
Affiliation(s)
- Kaiyuan Yu
- The Second Affiliated Hospital & YuYing Children’s Hospital of Wenzhou Medical University, Wenzhou City, People’s Republic of China
| | - Luxia Xiang
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Shaoxun Li
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Shuaibin Wang
- The Second Affiliated Hospital & YuYing Children’s Hospital of Wenzhou Medical University, Wenzhou City, People’s Republic of China
| | - Chaohao Chen
- The Second Affiliated Hospital & YuYing Children’s Hospital of Wenzhou Medical University, Wenzhou City, People’s Republic of China
| | - Haiqi Mu
- The Second Affiliated Hospital & YuYing Children’s Hospital of Wenzhou Medical University, Wenzhou City, People’s Republic of China
| |
Collapse
|
22
|
Cui J, Wang L, Ren X, Zhang Y, Zhang H. LRPPRC: A Multifunctional Protein Involved in Energy Metabolism and Human Disease. Front Physiol 2019; 10:595. [PMID: 31178748 PMCID: PMC6543908 DOI: 10.3389/fphys.2019.00595] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Accepted: 04/26/2019] [Indexed: 12/26/2022] Open
Abstract
The pentatricopeptide repeat (PPR) family plays a major role in RNA stability, regulation, processing, splicing, translation, and editing. Leucine-rich PPR-motif-containing protein (LRPPRC), a member of the PPR family, is a known gene mutation that causes Leigh syndrome French-Canadian. Recently, growing evidence has pointed out that LRPPRC dysregulation is related to various diseases ranging from tumors to viral infections. This review presents available published data on the LRPPRC protein function and its role in tumors and other diseases. As a multi-functional protein, LRPPRC regulates a myriad of biological processes, including energy metabolism and maturation and the export of nuclear mRNA. Overexpression of LRPPRC has been observed in various human tumors and is associated with poor prognosis. Downregulation of LRPPRC inhibits growth and invasion, induces apoptosis, and overcomes drug resistance in tumor cells. In addition, LRPPRC plays a potential role in Parkinson's disease, neurofibromatosis 1, viral infections, and venous thromboembolism. Further investigating these new functions of LRPPRC should provide novel opportunities for a better understanding of its pathological role in diseases from tumors to viral infections and as a potential biomarker and molecular target for disease treatment.
Collapse
Affiliation(s)
- Jie Cui
- Department of Oncology, The First Affiliated Hospital, Xi'an Medical University, Xi'an, China.,College of General Practitioners, Xi'an Medical University, Xi'an, China
| | - Li Wang
- Department of Oncology, The First Affiliated Hospital, Xi'an Medical University, Xi'an, China.,College of General Practitioners, Xi'an Medical University, Xi'an, China
| | - Xiaoyue Ren
- Department of Oncology, The First Affiliated Hospital, Xi'an Medical University, Xi'an, China.,College of General Practitioners, Xi'an Medical University, Xi'an, China
| | - Yamin Zhang
- Department of Oncology, The First Affiliated Hospital, Xi'an Medical University, Xi'an, China.,College of General Practitioners, Xi'an Medical University, Xi'an, China
| | - Hongyi Zhang
- College of General Practitioners, Xi'an Medical University, Xi'an, China.,Department of Urology, The First Affiliated Hospital, Xi'an Medical University, Xi'an, China
| |
Collapse
|
23
|
Jaber FA, Khan NM, Ansari MY, Al-Adlaan AA, Hussein NJ, Safadi FF. Autophagy plays an essential role in bone homeostasis. J Cell Physiol 2019; 234:12105-12115. [PMID: 30820954 DOI: 10.1002/jcp.27071] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 06/29/2018] [Indexed: 12/20/2022]
Abstract
Autophagy is very critical for multiple cellular processes. Autophagy plays a critical role in bone cell differentiation and function.
Collapse
Affiliation(s)
- Fatima A Jaber
- Department of Biology, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Anatomy and Neurobiology, Northeast Ohio Medical University (NEOMED) School of Medicine, Rootstown, Ohio.,School of Biomedical Sciences, Kent State University, Kent, Ohio
| | - Nazir M Khan
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University (NEOMED) School of Medicine, Rootstown, Ohio
| | - Mohammad Y Ansari
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University (NEOMED) School of Medicine, Rootstown, Ohio
| | - Asaad A Al-Adlaan
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University (NEOMED) School of Medicine, Rootstown, Ohio.,School of Biomedical Sciences, Kent State University, Kent, Ohio
| | - Nazar J Hussein
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University (NEOMED) School of Medicine, Rootstown, Ohio.,School of Biomedical Sciences, Kent State University, Kent, Ohio
| | - Fayez F Safadi
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University (NEOMED) School of Medicine, Rootstown, Ohio.,School of Biomedical Sciences, Kent State University, Kent, Ohio.,Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, Ohio.,Department of Orthopedic Surgery, SUMMA Health System, Akron, Ohio.,Rebecca D. Considine Research Institute Akron Children's Hospital, Akron, Ohio
| |
Collapse
|
24
|
Ricci M, Frantellizzi V, Bulzonetti N, De Vincentis G. Reversibility of castration resistance status after Radium-223 dichloride treatment: clinical evidence and review of the literature. Int J Radiat Biol 2019; 95:554-561. [PMID: 30557063 DOI: 10.1080/09553002.2019.1558301] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In the history of prostate cancer, some of the patients progressed to castration-resistant prostate cancer (CRPC) stage and, although new drugs and treatment protocols have been introduced, CRPC presents poor prognosis. This review is focused on biological mechanisms, underlying CRPC described in scientific literature in order to explain the reversion of resistance to castration. We present the case of a 73-year-old man, affected by bone metastatic CRPC, early treated with Radium-223 with a complete response. After 15 months from Radium-223 treatment, prostate-specific antigen increased with radiological progression. Androgen deprivation therapy was again performed and was effective, despite previous CRPC condition and no known mechanisms that may explain the reversion of this condition. Therefore, to our knowledge, he is the unique described case of the reversion of resistance to castration. Nevertheless, promising aspects may be lack of intrametastatic production of androgen or the suppression of bypass androgen receptor signaling pathways. Furthermore, the cytotoxic action of Radium-223 on cancer stem cell (CSC), due to surrounding clones with high-bone turnover, or the immune response that underlying the abscopal effect, may also modulate the reversion of CRPC after Radium-223. If confirmed by multicenter trials, the reversion of CRPC may impact on the management of prostate cancer.
Collapse
Affiliation(s)
- Maria Ricci
- a Department of Radiological Sciences, Oncology and Anatomical Pathology , Sapienza University of Rome , Rome , Italy
| | - Viviana Frantellizzi
- a Department of Radiological Sciences, Oncology and Anatomical Pathology , Sapienza University of Rome , Rome , Italy.,b PhD Program: Angio-Cardio-Thoracic Pathophisiology and Imaging , "Sapienza" University of Rome , Rome , Italy
| | - Nadia Bulzonetti
- c Department of Radiotherapy , Policlinico Umberto I, Sapienza University of Rome , Rome , Italy
| | - Giuseppe De Vincentis
- a Department of Radiological Sciences, Oncology and Anatomical Pathology , Sapienza University of Rome , Rome , Italy
| |
Collapse
|
25
|
Huang Y, Jiang G, Liang X, Lan Z, Su Z, Wu H, Weng J, Jiang X. Elevated expression of PTCD3 correlates with tumor progression and predicts poor prognosis in patients with prostate cancer. Mol Med Rep 2018; 18:3914-3922. [PMID: 30132530 PMCID: PMC6131642 DOI: 10.3892/mmr.2018.9402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 08/06/2018] [Indexed: 11/23/2022] Open
Abstract
Pentatricopeptide repeat domain protein 3 (PTCD3) is a mitochondrial RNA-binding protein that serves a role in mitochondrial translation. PTCD3 was originally reported as an oncogene that is involved in breast cancer and lymphoma. However, the expression and function of PTCD3 in prostate cancer (PCa) are unknown. Therefore, the aim of the present study was to investigate the expression of PTCD3 and its clinical significance in PCa. Immunohistochemistry and dataset analyses revealed that PTCD3 protein expression levels were enhanced in human PCa tissues and mouse PCa models. PTCD3 expression levels were positively correlated with advanced PCa pathological grade and clinical stage. Additionally, PTCD3 mRNA expression was positively correlated with tissue malignancy, high Gleason score and distant metastasis in The Cancer Genome Atlas dataset. Kaplan-Meier analysis revealed that high PTCD3 levels can predict the increased biochemical recurrence (BCR)-free survival in all patients with or without metastasis. The overexpression of PTCD3 could be used as an independent prognostic marker of poor BCR-free survival. Immunofluorescence and western blot analysis in human PCa cell lines further confirmed that PTCD3 levels were associated with the hormone independence of PCa. Therefore, the present study revealed that PTCD3 levels may serve as a novel biomarker for PCa prognosis.
Collapse
Affiliation(s)
- Yiqiao Huang
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| | - Ganggang Jiang
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| | - Xue Liang
- Department of Central Laboratory, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| | - Ziquan Lan
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| | - Zhengming Su
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| | - Hualing Wu
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| | - Jinsheng Weng
- Department of Central Laboratory, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| | - Xianhan Jiang
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| |
Collapse
|
26
|
Parousis A, Carter HN, Tran C, Erlich AT, Mesbah Moosavi ZS, Pauly M, Hood DA. Contractile activity attenuates autophagy suppression and reverses mitochondrial defects in skeletal muscle cells. Autophagy 2018; 14:1886-1897. [PMID: 30078345 PMCID: PMC6152519 DOI: 10.1080/15548627.2018.1491488] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 06/05/2018] [Accepted: 06/18/2018] [Indexed: 12/18/2022] Open
Abstract
Macroautophagy/autophagy is a survival mechanism that facilitates protein turnover in post-mitotic cells in a lysosomal-dependent process. Mitophagy is a selective form of autophagy, which arbitrates the selective recognition and targeting of aberrant mitochondria for degradation. Mitochondrial content in cells is the net balance of mitochondrial catabolism via mitophagy, and organelle biogenesis. Although the latter process has been well described, mitophagy in skeletal muscle is less understood, and it is currently unknown how these two opposing mechanisms converge during contractile activity. Here we show that chronic contractile activity (CCA) in muscle cells induced mitochondrial biogenesis and coordinately enhanced the expression of TFEB (transcription factor EB) and PPARGC1A/PGC-1α, master regulators of lysosome and mitochondrial biogenesis, respectively. CCA also enhanced the expression of PINK1 and the lysosomal protease CTSD (cathepsin D). Autophagy blockade with bafilomycin A1 (BafA) reduced mitochondrial state 3 and 4 respiration, increased ROS production and enhanced the accumulation of MAP1LC3B-II/LC3-II and SQSTM1/p62. CCA ameliorated this mitochondrial dysfunction during defective autophagy, increased PPARGC1A, normalized LC3-II levels and reversed mitochondrially-localized SQSTM1 toward control levels. NAC emulated the LC3-II reductions induced by contractile activity, signifying that a decrease in oxidative stress could represent a mechanism of autophagy normalization brought about by CCA. CCA enhances mitochondrial biogenesis and lysosomal activity, and normalizes autophagy flux during autophagy suppression, partly via ROS-dependent mechanisms. Thus, contractile activity represents a potential therapeutic intervention for diseases in which autophagy is inhibited, such as vacuolar myopathies in skeletal muscle, by establishing a healthy equilibrium of anabolic and catabolic pathways. ABBREVIATIONS AMPK: AMP-activated protein kinase; BafA: bafilomycin A1; BNIP3L: BCL2/adenovirus E1B interacting protein 3-like; CCA: chronic contractile activity; COX4I1: cytochrome c oxidase subunit 4I1; DMEM: Dulbecco's modified Eagle's medium; GFP: green fluorescent protein; LSD: lysosomal storage diseases; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; MTORC1: mechanistic target of rapamycin kinase complex 1; NAC: N-acetylcysteine; PPARGC1A: peroxisome proliferative activated receptor, gamma, coactivator 1 alpha; PINK1: PTEN induced putative kinase 1; ROS: reactive oxygen species; SOD2: superoxide dismutase 2, mitochondrial; SQSTM1/p62: sequestosome 1; TFEB: transcription factor EB.
Collapse
Affiliation(s)
- Alexa Parousis
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - Heather N. Carter
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - Claudia Tran
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - Avigail T. Erlich
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - Zahra S. Mesbah Moosavi
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - Marion Pauly
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - David A. Hood
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| |
Collapse
|
27
|
Jiang X, Huang Y, Liang X, Jiang F, He Y, Li T, Xu G, Zhao H, Yang W, Jiang G, Su Z, Jiang L, Liu L. Metastatic prostate cancer-associated P62 inhibits autophagy flux and promotes epithelial to mesenchymal transition by sustaining the level of HDAC6. Prostate 2018; 78:426-434. [PMID: 29383752 PMCID: PMC5897115 DOI: 10.1002/pros.23487] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 01/05/2018] [Indexed: 12/28/2022]
Abstract
BACKGROUND P62 (also named sequestosome-1, SQSTM1) is involved in autophagy regulation through multiple pathways. It interacts with autophagosomes-associated LC3-II and ubiquitinated protein aggregates to engulf the aggregates in autophagosomes, interacts with HDAC6 to inhibit its deacetylase activity to maintain the levels of acetylated α-tubulin and stabilities of microtubules to enhance autophagosome trafficking, and regulates autophagy initiation and cell survival. We performed immunohistochemistry staining of P62 in prostate tissues from prostate cancer patients and found that levels of P62 in patients with prostate adenocarcinomas (PCA) are significantly higher than those in patients with benign prostate hyperplasia (BPH). High levels of P62 predict high tumor grade and high intensity of metastasis. METHODS We created prostate cancer cell lines stably overexpressing P62 and then suppress the expression of P62 in the cell line stably overexpressing P62 with CRISPR technology. Cell proliferation assay with crystal violet, cell migration assay, cell invasion assay, Western blot analysis, and confocal fluorescent microscopy were conducted to test the impact of altered levels of P62 on the growth, migration, invasion, epithelial-to-mesenchymal transition, autophagy flux, HDAC6 activity, and microtubular acetylation of cancer cells. RESULTS P62 increased the levels of HDAC6 and reduced the acetylation of α-tubulin and the stability of microtubules. Consequently, high levels of P62 caused a promotion of epithelial-to-mesenchymal transition in addition to an impairment of autophagy flux, and further led to an enhancement of proliferation, migration, and invasion of prostate cancer cells. CONCLUSION P62 promotes metastasis of PCA by sustaining the level of HDAC6 to inhibit autophagy and promote epithelial-to-mesenchymal transition.
Collapse
Affiliation(s)
- Xianhan Jiang
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yiqiao Huang
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xue Liang
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Funeng Jiang
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yongzhong He
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Tian Li
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Guibin Xu
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Haibo Zhao
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Weiqing Yang
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ganggang Jiang
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhengming Su
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lingke Jiang
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Leyuan Liu
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Texas A&M University, Houston, Texas, USA
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University, College Station, Texas, USA
- Correspondence to Leyuan Liu, PhD, Center for Translational Cancer Research, Texas A&M Institute of Biosciences and Technology, Texas A&M University, 2121 W. Holcombe Blvd., Houston, Texas, 77030, USA.
| |
Collapse
|
28
|
Huang H, Wang Q, Du T, Lin C, Lai Y, Zhu D, Wu W, Ma X, Bai S, Li Z, Liu L, Li Q. Matrine inhibits the progression of prostate cancer by promoting expression of GADD45B. Prostate 2018; 78:327-335. [PMID: 29356020 DOI: 10.1002/pros.23469] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 11/29/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND Matrine is a naturally occurring alkaloid extracted from the Chinese herb Sophora flavescens. It has been demonstrated to exhibit antiproliferative properties, promote apoptosis, and inhibit cell invasion in a number of cancer cell lines by modulating the NF-κB pathway to downregulate the expression of MMP2 and MM9. It has also been shown to improve the efficacy of chemotherapy when it is combined with other chemotherapy drugs. However, the therapeutic potential of matrine for prostate cancer needs to be further studied. METHODS We analyzed KEGG pathways of differential gene expression between matrine-treated and untreated prostate cancer cell lines and identified GADD45B as one of major target genes of matrine based on its role in apoptosis and prognosis value for prostate cancer patients in TCGA database. We further analyzed the expression of GADD45B protein in a tissue microarray and mRNA in TCGA database, and tested the synergistic impacts of matrine and GADD45B overexpression on proliferation, apoptosis, migration and invasion of prostate cancer cell DU145. RESULTS Matrine promoted the expression of GADD45B, a tumor suppressive gene that is involved in the regulation of cell cycle, DNA damage repair, cell survival, aging, apoptosis and other cellular processes through p38/JNK, ROS-GADD45B-p38, or other signal pathways. Although GADD45B is elevated in prostate cancer tissues, levels of GADD45B in prostate tumor tissues are reduced at late stage of tumor invasion, and higher levels of GADD45B predict better survivals of prostate cancer patients. CONCLUSIONS Matrine may be used to treat prostate cancer patients to increase the levels of GADD45B to inhibit tumor invasion and improve patient survivals.
Collapse
Affiliation(s)
- Hai Huang
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Center for Translational Cancer Research, Texas A&M Institute of Biosciences and Technology, Texas A&M University, Houston, Texas
| | - Qiong Wang
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Tao Du
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Department of Obstetrics and Gynecology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Chunhao Lin
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yiming Lai
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Dingjun Zhu
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Wanhua Wu
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiaoming Ma
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Soumin Bai
- Department of Radiation Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zean Li
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Leyuan Liu
- Center for Translational Cancer Research, Texas A&M Institute of Biosciences and Technology, Texas A&M University, Houston, Texas
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University, College Station, Texas
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qi Li
- Department of Clinical Laboratory, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
29
|
Huang Y, Jiang X, Liang X, Jiang G. Molecular and cellular mechanisms of castration resistant prostate cancer. Oncol Lett 2018; 15:6063-6076. [PMID: 29616091 PMCID: PMC5876469 DOI: 10.3892/ol.2018.8123] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Accepted: 01/26/2018] [Indexed: 12/21/2022] Open
Abstract
With increases in the mortality rate and number of patients with prostate cancer (PCa), PCa, particularly the advanced and metastatic disease, has been the focus of a number of studies globally. Over the past seven decades, androgen deprivation therapy has been the primary therapeutic option for patients with advanced PCa; however, the majority of patients developed a poor prognosis stage of castration resistant prostate cancer (CRPC), which eventually led to mortality. Due to CRPC being incurable, laboratory investigations and clinical studies focusing on CRPC have been conducted worldwide. Clarification of the molecular pathways that may lead to CRPC is important for discovering novel therapeutic strategies to delay or reverse the progression of disease. A sustained androgen receptor (AR) signal is still regarded as the main cause of CRPC. Increasing number of studies have proposed different potential mechanisms that cause CRPC, and this has led to the development of novel agents targeting the AR-dependent pathway or AR-independent signaling. In the present review, the major underlying mechanisms causing CRPC, including several major categories of AR-dependent mechanisms, AR bypass signaling, AR-independent mechanisms and other important hypotheses (including the functions of autophagy, PCa stem cell and microRNAs in CRPC progression), are summarized with retrospective pre-clinical or clinical trials to guide future research and therapy.
Collapse
Affiliation(s)
- Yiqiao Huang
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| | - Xianhan Jiang
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| | - Xue Liang
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| | - Ganggang Jiang
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| |
Collapse
|
30
|
Nishio T, Kurabe N, Goto-Inoue N, Nakamura T, Sugimura H, Setou M, Maekawa M. Immunohistochemical expression analysis of leucine-rich PPR-motif-containing protein (LRPPRC), a candidate colorectal cancer biomarker identified by shotgun proteomics using iTRAQ. Clin Chim Acta 2017; 471:276-282. [PMID: 28622966 DOI: 10.1016/j.cca.2017.06.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 05/30/2017] [Accepted: 06/12/2017] [Indexed: 11/16/2022]
Abstract
BACKGROUND Colorectal cancer (CRC) is the fourth most frequent cause of cancer deaths in the world. Novel biomarkers for the diagnosis, prognosis, and treatment of CRC are required to improve the clinical strategy. METHODS We applied shotgun proteomics using isobaric tags for relative and absolute quantitation (iTRAQ) to identify novel biomarkers of CRC, and then we detected leucine-rich PPR-motif-containing protein (LRPPRC) expression in 83 normal colorectal tissues and 133 CRC tissues by immunohistochemistry. RESULTS A total of 570 proteins were identified using iTRAQ. We validated the expression of LRPPRC protein by immunohistochemical analysis of the 77 proteins that showed expression changes in the cancer tissues >1.5-fold the levels in the normal tissues. The expression levels of LRPPRC were significantly higher in CRC tissues than those in normal colorectal tissues, and the expression levels were related with tumor differentiation and especially high in moderately differentiated CRC tissues. CONCLUSION We identified a novel, differentially expressed protein, LRPPRC, which has the potential to serve as a molecular target for diagnosis and/or prognosis of CRC.
Collapse
Affiliation(s)
- Tomohisa Nishio
- Department of Laboratory Medicine, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu 431-3192, Japan; Department of Cell Biology and Anatomy, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu 431-3192, Japan; Tsukuba Research Institute, Sekisui Medical Co., Ltd., 3-3-1, Kouyoudai, Ryugasaki 301-0852, Japan
| | - Nobuya Kurabe
- Department of Tumor Pathology, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu 431-3192, Japan
| | - Naoko Goto-Inoue
- Department of Cell Biology and Anatomy, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu 431-3192, Japan
| | - Toshio Nakamura
- Second Department of Surgery, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu 431-3192, Japan; Department of Surgery, Fujieda Municipal General Hospital, 4-1-11, Surugadai, Fujieda 426-8677, Japan
| | - Haruhiko Sugimura
- Department of Tumor Pathology, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu 431-3192, Japan
| | - Mitsutoshi Setou
- Department of Cell Biology and Anatomy, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu 431-3192, Japan
| | - Masato Maekawa
- Department of Laboratory Medicine, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu 431-3192, Japan.
| |
Collapse
|
31
|
Mitochondrion-associated protein peroxiredoxin 3 promotes benign prostatic hyperplasia through autophagy suppression and pyroptosis activation. Oncotarget 2017; 8:80295-80302. [PMID: 29113303 PMCID: PMC5655198 DOI: 10.18632/oncotarget.17927] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Accepted: 05/03/2017] [Indexed: 12/26/2022] Open
Abstract
Benign prostatic hyperplasia (BPH) is one of the most common diseases in the senior men and age plays an important role in the initiation and development of BPH. Mammalian cells primarily use the autophagy-lysosome system to degrade misfolded/aggregated proteins and dysfunctional organelles such as mitochondria and suppress pyroptosis, a type of cell death that stimulates inflammatory responses and growth of other cells around. Peroxiredoxin 3 (PRDX3) is the only mitochondrion-associated member of peroxiredoxin family enzymes that exert their protective antioxidant role in cells through their peroxidase activity. We hypothesized that PRDX3 may inhibit autophagy to activate pyroptosis to induce growth of prostatic epithelial cells. Here we show that PRDX3 maintained the integrity of mitochondria and its depletion led to an enhancement of oxidative stresses. PRDX3-associated and PRDX3-free mitochondria co-existed in the same cells. PRDX3 expressed at higher levels in prostatic epithelial cells in prostate tissues from BPH patients and BPH-representative cell line than in prostate tissues from healthy donors and a cell line representing normal epithelial cells. PRDX3 suppressed autophagy flux and activated pyroptosis to induce inflammatory responses and stimulate the over-growth of prostate tissues. Therefore, higher levels of PDRX3 in prostatic epithelial cells may promote the initiation and development of BPH through autophagy inhibition and pyroptosis activation.
Collapse
|
32
|
Huang H, Du T, Zhang Y, Lai Y, Li K, Fan X, Zhu D, Lin T, Xu K, Huang J, Liu L, Guo Z. Elevation of SHARPIN Protein Levels in Prostate Adenocarcinomas Promotes Metastasis and Impairs Patient Survivals. Prostate 2017; 77:718-728. [PMID: 28230260 DOI: 10.1002/pros.23302] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 12/20/2016] [Indexed: 12/25/2022]
Abstract
BACKGROUND SHARPIN, SHANK-associated RH domain interacting protein, associates with a linear ubiquitin chain assembly complex (LUBAC) to regulate inflammation and immunity. It has been reported that SHARPIN is highly expressed in several human tumors including ovarian cancer and liver cancer. We found that SHARPIN is also highly expressed in prostate cancer cell lines of DU145, LNCAP, and PC-3. Suppression of SHARPIN caused an inhibition of NF-κB signal and decreases in tumorigenesis of cultured cells in NOD/SCID mouse model. Overexpression of SHARPIN in prostate cancer cells promoted cell growth and reduced apoptosis through NF-kB/ERK/Akt pathway and apoptosis-associated proteins. METHODS We analyzed the expression of SHARPIN in prostate cancer tissues from 95 patients and its relationship with other clinical characteristics associated with PCA malignancies and patient survivals, and examined the impacts of SHARPIN suppression with siRNA on proliferation, angiogenesis, invasion, and expression levels of MMP-9 of prostate cancer cells and metastasis to lung by these cells in nude mice. RESULTS High levels of SHARPIN were associated with high malignancies of PCA and predicted shorter survivals of PCA patients. Suppression of SHARPIN impaired cell proliferation, angiogenesis, and invasion and reduced levels of MMP-9 in prostate cancer cells and reduced the size of metastatic lung tumors induced by these cells in mice. CONCLUSIONS SHARPIN enhances the metastasis of prostate cancer and impair patient survivals. Prostate 77:718-728, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Hai Huang
- Department of Urology, The Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M University, Houston, Texas
| | - Tao Du
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M University, Houston, Texas
- Department of Gynecology & Obstetrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yiming Zhang
- Department of Urology, The Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yiming Lai
- Department of Urology, The Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kaiwen Li
- Department of Urology, The Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xinxing Fan
- Department of Urology, The Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dingjun Zhu
- Department of Urology, The Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tianxin Lin
- Department of Urology, The Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Kewei Xu
- Department of Urology, The Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jian Huang
- Department of Urology, The Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Leyuan Liu
- Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M University, Houston, Texas
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University, College Station, Houston, Texas
| | - Zhenghui Guo
- Department of Urology, The Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
33
|
Yue F, Li W, Zou J, Jiang X, Xu G, Huang H, Liu L. Spermidine Prolongs Lifespan and Prevents Liver Fibrosis and Hepatocellular Carcinoma by Activating MAP1S-Mediated Autophagy. Cancer Res 2017; 77:2938-2951. [PMID: 28386016 DOI: 10.1158/0008-5472.can-16-3462] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 01/31/2017] [Accepted: 03/31/2017] [Indexed: 12/23/2022]
Abstract
Liver fibrosis and hepatocellular carcinoma (HCC) have worldwide impact but continue to lack safe, low cost, and effective treatments. In this study, we show how the simple polyamine spermidine can relieve cancer cell defects in autophagy, which trigger oxidative stress-induced cell death and promote liver fibrosis and HCC. We found that the autophagic marker protein LC3 interacted with the microtubule-associated protein MAP1S, which positively regulated autophagy flux in cells. MAP1S stability was regulated in turn by its interaction with the histone deacetylase HDAC4. Notably, MAP1S-deficient mice exhibited a 20% reduction in median survival and developed severe liver fibrosis and HCC under stress. Wild-type mice or cells treated with spermidine exhibited a relative increase in MAP1S stability and autophagy signaling via depletion of cytosolic HDAC4. Extending recent evidence that orally administered spermidine can extend lifespan in mice, we determined that life extension of up to 25% can be produced by lifelong administration, which also reduced liver fibrosis and HCC foci as induced by chemical insults. Genetic investigations established that these observed impacts of oral spermidine administration relied upon MAP1S-mediated autophagy. Our findings offer a preclinical proof of concept for the administration of oral spermidine to prevent liver fibrosis and HCC and potentially extend lifespan. Cancer Res; 77(11); 2938-51. ©2017 AACR.
Collapse
Affiliation(s)
- Fei Yue
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, Texas
| | - Wenjiao Li
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, Texas
| | - Jing Zou
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, Texas
| | - Xianhan Jiang
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, Texas.,The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Guibin Xu
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, Texas.,The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Hai Huang
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, Texas
| | - Leyuan Liu
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, Texas. .,The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China.,Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University, College Station, Texas
| |
Collapse
|
34
|
Xu G, Jiang Y, Xiao Y, Liu XD, Yue F, Li W, Li X, He Y, Jiang X, Huang H, Chen Q, Jonasch E, Liu L. Fast clearance of lipid droplets through MAP1S-activated autophagy suppresses clear cell renal cell carcinomas and promotes patient survival. Oncotarget 2017; 7:6255-65. [PMID: 26701856 PMCID: PMC4868754 DOI: 10.18632/oncotarget.6669] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 12/07/2015] [Indexed: 11/25/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is composed of cells whose cytoplasm filled with lipid droplets, subcellular organelles coated with adipocyte differentiation-related protein (ADFP) for the storage of triacylglycerol converted from excess free fatty acids. Mammalian cells primarily use the autophagy-lysosome system to degrade misfolded/aggregated proteins and dysfunctional organelles such as lipid droplets. MAP1S (originally named C19ORF5) is an autophagy activator and promotes the biogenesis and degradation of autophagosomes. Previously, we reported that MAP1S suppresses hepatocellular carcinogenesis in a mouse model and promoted the survival of patients with prostate adenocarcinomas by increasing the degradation of aggregated proteins and dysfunctional mitochondria. Here we show that a suppression of MAP1S in renal cells causes an impairment of autophagic clearance of lipid droplets. In contrast, an overexpression of MAP1S causes an activation of autophagy flux and a reduction of lipid droplets so less DNA double strand breakage is induced. The levels of MAP1S in normal renal cells are dramatically higher than those in the ccRCC tissues and cell lines derived from renal cell carcinomas. High levels of MAP1S are associated with a reduced malignancy and metastasis of ccRCC and predict a better survival of ccRCC patients. Therefore, autophagy defects in the degradation of lipid droplets triggered by the MAP1S deficiency may enhance the initiation and development of ccRCC and reduce the survival of ccRCC patients.
Collapse
Affiliation(s)
- Guibin Xu
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, 510700, China.,Center for The Innovation and Translation of Minimally Invasive Techniques, Guangzhou Medical University, Guangzhou, Guangdong Province, 510700, China.,Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, 77030, USA
| | - Yaodong Jiang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| | - Yuansong Xiao
- Department of Urology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, Guangdong Province, 510010, China
| | - Xian D Liu
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Fei Yue
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, 77030, USA
| | - Wenjiao Li
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, 77030, USA
| | - Xun Li
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, 510700, China.,Center for The Innovation and Translation of Minimally Invasive Techniques, Guangzhou Medical University, Guangzhou, Guangdong Province, 510700, China
| | - Yongzhong He
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, 510700, China.,Center for The Innovation and Translation of Minimally Invasive Techniques, Guangzhou Medical University, Guangzhou, Guangdong Province, 510700, China
| | - Xianhan Jiang
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, 510700, China.,Center for The Innovation and Translation of Minimally Invasive Techniques, Guangzhou Medical University, Guangzhou, Guangdong Province, 510700, China
| | - Hai Huang
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, 77030, USA
| | - Qi Chen
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, 77030, USA
| | - Eric Jonasch
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Leyuan Liu
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, 77030, USA.,Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M Health Science Center, College Station, Texas, 77843, USA
| |
Collapse
|
35
|
Zhang HY, Ma YD, Zhang Y, Cui J, Wang ZM. Elevated levels of autophagy-related marker ULK1 and mitochondrion-associated autophagy inhibitor LRPPRC are associated with biochemical progression and overall survival after androgen deprivation therapy in patients with metastatic prostate cancer. J Clin Pathol 2016; 70:383-389. [PMID: 27679555 DOI: 10.1136/jclinpath-2016-203926] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 09/01/2016] [Accepted: 09/04/2016] [Indexed: 11/03/2022]
Abstract
AIM To evaluate the expression levels and prognostic significance of autophagy-related markers, UNC-51-like kinase1 (ULK1), Beclin1, microtubule-associated protein light chain 3 (LC3), autophagy-related gene 5 (ATG5) and mitochondrion-associated autophagy inhibitor, LRPPRC, in patients with metastatic prostate cancer (PCa) after androgen deprivation therapy (ADT). METHODS Expressions of ULK1, Beclin1, LC3, ATG5 and LRPPRC were assessed by immunohistochemical examination in 198 patients with metastatic PCa who were receiving ADT (goserelin and bicalutamide). RESULTS High expression levels of LRPPRC and ULK1were significantly associated with Gleason score, serum prostate-specific antigen (PSA) levels, PSA levels after ADT and number of metastatic sites. High expression of ULK1 in patients with concomitant high expression of LRPPRC was significantly associated with multiple metastases, shorter biochemical progression (BCP)-free survival and shorter overall survival (OS). ULK1 expression, LRPPRC expression, Gleason score, PSA levels after ADT and number of metastatic sites were independently associated with shorter BCP-free survival and OS on multivariate analysis. Furthermore, two-year BCP rate of patients with ≥3 risk factors was found to be significantly higher as compared with that of patients with ≤1 and 2 risk factors. Three-year OS rate in patients with ≥3 risk factors was significantly lower than that of those with ≤1 and 2 risk factors. CONCLUSIONS High expression of ULK1 concomitant with high expression of LRPPRC may serve as useful markers for shorter BCP-free survival and OS in patients with metastatic PCa after ADT.
Collapse
Affiliation(s)
- Hong-Yi Zhang
- Department of Urology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China.,Department of Urology, Yanan University Affiliated Hospital, Yan'an, Shaanxi Province, People's Republic of China
| | - Ya-Dong Ma
- Department of Urology, Yanan University Affiliated Hospital, Yan'an, Shaanxi Province, People's Republic of China
| | - Ye Zhang
- Department of Urology, Yanan University Affiliated Hospital, Yan'an, Shaanxi Province, People's Republic of China
| | - Jie Cui
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
| | - Zi-Ming Wang
- Department of Urology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
| |
Collapse
|
36
|
Li W, Zou J, Yue F, Song K, Chen Q, McKeehan WL, Wang F, Xu G, Huang H, Yi J, Liu L. Defects in MAP1S-mediated autophagy cause reduction in mouse lifespans especially when fibronectin is overexpressed. Aging Cell 2016; 15:370-9. [PMID: 26750654 PMCID: PMC4783353 DOI: 10.1111/acel.12441] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2015] [Indexed: 02/02/2023] Open
Abstract
Autophagy is a cellular process that executes the turnover of dysfunctional organelles and misfolded or abnormally aggregated proteins. Microtubule‐associated protein MAP1S interacts with autophagy marker LC3 and positively regulates autophagy flux. LC3 binds with fibronectinmRNA and facilitates its translation. The synthesized fibronectin protein is exported to cell surface to initiate the assembly of fibronectin extracellular matrix. Fibronectin is degraded in lysosomes after it is engulfed into cytosol via endocytosis. Here, we show that defects in MAP1S‐mediated autophagy trigger oxidative stress, sinusoidal dilation, and lifespan reduction. Overexpression of LC3 in wild‐type mice increases the levels of fibronectin and γ‐H2AX, a marker of DNA double‐strand breakage. LC3‐induced fibronectin is efficiently degraded in lysosomes to maintain a balance of fibronectin levels in wild‐type mice so that the mice live a normal term of lifespan. In the LC3 transgenic mice with MAP1S deleted, LC3 enhances the synthesis of fibronectin but the MAP1S depletion causes an impairment of the lysosomal degradation of fibronectin. The accumulation of fibronectin protein promotes liver fibrosis, induces an accumulation of cell population at the G0/G1 stage, and further intensifies oxidative stress and sinusoidal dilatation. The LC3‐induced overexpression of fibronectin imposes stresses on MAP1S‐deficient mice and dramatically reduces their lifespans. Therefore, MAP1S‐mediated autophagy plays an important role in maintaining mouse lifespan especially in the presence of extra amount of fibronectin.
Collapse
Affiliation(s)
- Wenjiao Li
- Center for Translational Cancer Research Institute of Biosciences and Technology Texas A&M Health Science Center 2121 W. Holcombe Blvd. Houston TX 77030 USA
| | - Jing Zou
- Center for Translational Cancer Research Institute of Biosciences and Technology Texas A&M Health Science Center 2121 W. Holcombe Blvd. Houston TX 77030 USA
- Department of ophthalmology Xiangya Hospital Central South University Changsha Hunan 410008 China
- Jiangxi Research Institute of Ophthalmology and Visual Sciences The Affiliated Eye Hospital of Nanchang University Nanchang 330006 China
| | - Fei Yue
- Center for Translational Cancer Research Institute of Biosciences and Technology Texas A&M Health Science Center 2121 W. Holcombe Blvd. Houston TX 77030 USA
| | - Kun Song
- Center for Translational Cancer Research Institute of Biosciences and Technology Texas A&M Health Science Center 2121 W. Holcombe Blvd. Houston TX 77030 USA
| | - Qi Chen
- Center for Translational Cancer Research Institute of Biosciences and Technology Texas A&M Health Science Center 2121 W. Holcombe Blvd. Houston TX 77030 USA
| | - Wallace L. McKeehan
- Center for Translational Cancer Research Institute of Biosciences and Technology Texas A&M Health Science Center 2121 W. Holcombe Blvd. Houston TX 77030 USA
| | - Fen Wang
- Center for Translational Cancer Research Institute of Biosciences and Technology Texas A&M Health Science Center 2121 W. Holcombe Blvd. Houston TX 77030 USA
| | - Guibin Xu
- Center for Translational Cancer Research Institute of Biosciences and Technology Texas A&M Health Science Center 2121 W. Holcombe Blvd. Houston TX 77030 USA
| | - Hai Huang
- Center for Translational Cancer Research Institute of Biosciences and Technology Texas A&M Health Science Center 2121 W. Holcombe Blvd. Houston TX 77030 USA
- Department of Urology The Sun Yat‐sen Memorial Hospital Sun Yat‐sen University Guangzhou 510120 China
| | - Jinglin Yi
- Jiangxi Research Institute of Ophthalmology and Visual Sciences The Affiliated Eye Hospital of Nanchang University Nanchang 330006 China
| | - Leyuan Liu
- Center for Translational Cancer Research Institute of Biosciences and Technology Texas A&M Health Science Center 2121 W. Holcombe Blvd. Houston TX 77030 USA
- Jiangxi Research Institute of Ophthalmology and Visual Sciences The Affiliated Eye Hospital of Nanchang University Nanchang 330006 China
- Department of Molecular and Cellular Medicine College of Medicine Texas A&M Health Science Center College Station TX 77843 USA
| |
Collapse
|
37
|
Stem cells, biomarkers and genetic profiling: approaching future challenges in Urology. Urologia 2016; 83:4-13. [PMID: 26940971 DOI: 10.5301/uro.5000165] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2016] [Indexed: 12/13/2022]
Abstract
Urological research is facing future challenges, the most difficult one is the fast and meaningful transfer of the massive amount of data from research basic to clinical practice. Between the most important issues that research should focus in the next years are targeting of tumor stem cells, clinical application of biomarkers, and wide application of genetic profiling of urological neoplasms. Several clinical implications are expected, from diagnosis to selection of candidates for different treatment modalities, to modulation of sequential treatment plans, to prognosis. A number of clinical trials based on research data from the hottest issues are in the pipeline. In this review, we will focus on new insights from recent work worlwide in urological research, with particular attention to high-risk nonmuscle-invasive and muscle-invasive bladder cancer, prostate cancer, and kidney cancer. Cancer care is moving towards a personalized approach in patient management. The most important issues in urological research point strongly in this direction and show an enormous potential for the rapid landing of Urology in the era of personalized medicine.
Collapse
|
38
|
Song K, Hu W, Yue F, Zou J, Li W, Chen Q, Yao Q, Sun W, Liu L. Transforming Growth Factor TGFβ Increases Levels of Microtubule-Associated Protein MAP1S and Autophagy Flux in Pancreatic Ductal Adenocarcinomas. PLoS One 2015; 10:e0143150. [PMID: 26571030 PMCID: PMC4646605 DOI: 10.1371/journal.pone.0143150] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 10/30/2015] [Indexed: 02/02/2023] Open
Abstract
Background and Aim Autophagy is a cellular process to regulate the turnover of misfolded/aggregated proteins or dysfunctional organelles such as damaged mitochondria. Microtubule-associated protein MAP1S (originally named C19ORF5) is a widely-distributed homologue of neuronal-specific MAP1A and MAP1B with which autophagy marker light chain 3 (LC3) was originally co-purified. MAP1S bridges autophagic components with microtubules and mitochondria through LC3 and positively regulates autophagy flux from autophagosomal biogenesis to degradation. The MAP1S-mediated autophagy suppresses tumorigenesis as suggested in a mouse liver cancer model and in prostate cancer patients. The TGFβ signaling pathway plays a central role in pancreatic tumorigenesis, and high levels of TGFβ suggest a tumor suppressive function and predict a better survival for some patients with resectable pancreatic ductal adenocarcinoma. In this study, we try to understand the relationship between TGFβ and MAP1S-mediated autophagy in pancreatic ductal adenocarcinoma. Methods We collected the tumor and its adjacent normal tissues from 33 randomly selected patients of pancreatic ductal adenocarcinomas to test the association between TGFβ and autophagy markers MAP1S and LC3. Then we tested the cause and effect relation between TGFβ and autophagy markers in cultured pancreatic cancer cell lines. Results Here we show that levels of TGFβ and autophagy markers MAP1S and LC3 are dramatically elevated in tumor tissues from patients with pancreatic ductal adenocarcinomas. TGFβ increases levels of MAP1S protein and enhances autophagy flux. Conclusion TGFβ may suppress the development of pancreatic ductal adenocarcinomas by enhancing MAP1S-mediated autophagy.
Collapse
Affiliation(s)
- Kun Song
- Department of General Surgery, Xiangya Hospital, Central South University, Xiangya Road 87, 410008, Changsha, Hunan Province, China
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, United States of America
| | - Wei Hu
- The First People’s Hospital, Lian Yungang City, Jiangsu Province, China
| | - Fei Yue
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, United States of America
| | - Jing Zou
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, United States of America
| | - Wenjiao Li
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, United States of America
| | - Qi Chen
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, United States of America
| | - Qizhi Yao
- Department of Surgery, Molecular Surgeon Research Center, Baylor College of Medicine, One Baylor Plaza, Houston, Texas, United States of America
| | - Weijia Sun
- Department of General Surgery, Xiangya Hospital, Central South University, Xiangya Road 87, 410008, Changsha, Hunan Province, China
- * E-mail: (WS); (LL)
| | - Leyuan Liu
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, United States of America
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M Health Science Center, College Station, Texas, United States of America
- * E-mail: (WS); (LL)
| |
Collapse
|
39
|
Zia A, Bhatti A, John P, Kiani AK. Data interpretation: deciphering the biological function of Type 2 diabetes associated risk loci. Acta Diabetol 2015; 52:789-800. [PMID: 25585593 DOI: 10.1007/s00592-014-0700-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 12/09/2014] [Indexed: 10/24/2022]
Abstract
AIMS Type 2 diabetes (T2D) is a complex multifactorial disorder with more than 40 loci associated with disease susceptibility. Most of these genome-wide significant loci reside in noncoding regions, it is important to decipher the potential regulatory function of these variants and to differentiate between true and tag signals. Nowadays, databases are being developed to study and predict the function of these associated variants, and RegulomeDB is one such database. METHODS We used RegulomeDB to analyze the potential function of the associated variants reported in five genome-wide association studies (GWAS) of T2D. RESULTS We investigated the 1,567 single nucleotide polymorphisms (SNPs) with 989 SNPs with a score of 1-6. Of those 989 SNPs, only 64 returned with RegulomeDB score <3 (evidence of regulatory function), and only four of these were GWAS significant SNPs (THADA/rs10203174, score = 1b; UBE2E2/rs7612463, score = 2a; ARAP1/rs1552224 and TP53INP1/rs8996852, score = 2b). But only 63 % of the annotated SNPs showed regulatory function that is an important limitation of the RegulomeDB as this database only provides information of few regulatory elements. CONCLUSION This study further supports that some of the noncoding GWAS variants are the true associations and not the tag ones. This study also proves the utility and importance of the RegulomeDB and other such databases. Although it is an extensive database of regulatory elements but has certain limitation due to utilization of only few types of regulatory elements and pathways.
Collapse
Affiliation(s)
- Asima Zia
- Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Sector H-12, Islamabad, Pakistan
| | | | | | | |
Collapse
|
40
|
Liu B, Miyake H, Nishikawa M, Tei H, Fujisawa M. Expression Profile of Autophagy-related Markers in Localized Prostate Cancer: Correlation With Biochemical Recurrence After Radical Prostatectomy. Urology 2015; 85:1424-30. [PMID: 25881865 DOI: 10.1016/j.urology.2015.03.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 02/01/2015] [Accepted: 03/06/2015] [Indexed: 01/05/2023]
Abstract
OBJECTIVE To evaluate the expression of multiple molecular markers involved in autophagy, a cellular degradation pathway for the clearance of damaged or superfluous proteins and organelles, in localized prostate cancer (PC) to clarify the prognostic significance of these markers in patients undergoing radical prostatectomy (RP). METHODS Expression levels of 5 autophagy markers, including autophagy-related gene 5, autophagy-related gene 9, Beclin1, microtubule-associated protein light chain 3, and UNC-51-like kinase 1 (ULK1), in RP specimens from 160 consecutive patients with clinically localized PC were measured by immunohistochemical staining. RESULTS Of these 5 markers, ULK1 expression was significantly correlated with the incidence of biochemical recurrence (BR). On univariate analysis, ULK1 expression, serum prostate-specific antigen level, pathologic stage, Gleason score, seminal vesicle invasion, and surgical margin status were identified as significant predictors of BR. All these significant factors except for seminal vesicle invasion were independently associated with BR on multivariate analysis. Furthermore, significant differences in BR-free survival according to the positive numbers of these 5 independent risk factors were noted, that is, BR occurred in 2 of 33 patients negative for risk factors (6.1%), 20 of 76 patients positive for 1 or 2 risk factors (26.3%), and 38 of 51 patients positive for ≥3 risk factors (74.5%). CONCLUSION Collectively, these findings suggest that measurement of expression levels of potential autophagy markers, particularly ULK1, in RP specimens, in addition to conventional parameters, may contribute to the accurate prediction of BR after RP for localized PC.
Collapse
Affiliation(s)
- Bing Liu
- Division of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hideaki Miyake
- Division of Urology, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - Masatomo Nishikawa
- Division of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hiromoto Tei
- Division of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masato Fujisawa
- Division of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
41
|
Zou J, Li W, Misra A, Yue F, Song K, Chen Q, Guo G, Yi J, Kimata JT, Liu L. The viral restriction factor tetherin prevents leucine-rich pentatricopeptide repeat-containing protein (LRPPRC) from association with beclin 1 and B-cell CLL/lymphoma 2 (Bcl-2) and enhances autophagy and mitophagy. J Biol Chem 2015; 290:7269-79. [PMID: 25631043 DOI: 10.1074/jbc.m114.627679] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Tetherin has been characterized as a key factor that restricts viral particles such as HIV and hepatitis C virus on plasma membranes, acts as a ligand of the immunoglobulin-like transcript 7 (ILT7) receptor in tumor cells, and suppresses antiviral innate immune responses mediated by human plasmacytoid dendritic cells. However, the normal cellular function of Tetherin without viral infection is unknown. Here we show that Tetherin not only serves as a substrate of autophagy but itself regulates the initiation of autophagy. Tetherin interacts with the autophagy/mitophagy suppressor LRPPRC and prevents LRPPRC from forming a ternary complex with Beclin 1 and Bcl-2 so that Beclin 1 is released to bind with PI3KCIII (class III PI3K) to activate the initiation of autophagy. Suppression of Tetherin leads to impairment of autophagy, whereas overexpression of Tetherin causes activation of autophagy. Under mitophagic stress, Tetherin is concentrated on mitochondria engulfed in autophagosomes. Tetherin plays a general role in the degradation of autophagosomes containing not only the symbiotic mitochondria but also, possibly, the infected virus. Therefore, Tetherin may enhance autophagy and mitophagy to suppress tumorigenesis, enhance innate immune responses, or prevent T cell apoptosis or pyroptosis.
Collapse
Affiliation(s)
- Jing Zou
- From the Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, 77030, the Jiangxi Research Institute of Ophthalmology and Visual Sciences, The Affiliated Eye Hospital of Nanchang University, Nanchang, 330006, China
| | - Wenjiao Li
- From the Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, 77030
| | - Anisha Misra
- the Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas 77030, and
| | - Fei Yue
- From the Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, 77030
| | - Kun Song
- From the Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, 77030
| | - Qi Chen
- From the Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, 77030
| | - Guanghua Guo
- the Jiangxi Research Institute of Ophthalmology and Visual Sciences, The Affiliated Eye Hospital of Nanchang University, Nanchang, 330006, China
| | - Jinglin Yi
- the Jiangxi Research Institute of Ophthalmology and Visual Sciences, The Affiliated Eye Hospital of Nanchang University, Nanchang, 330006, China
| | - Jason T Kimata
- the Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas 77030, and
| | - Leyuan Liu
- From the Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, 77030, the Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M Health Science Center, College Station, Texas 77843
| |
Collapse
|
42
|
Zhou J, Zhang F, Hou X, Zhang N. Downregulation of LRPPRC Induces Apoptosis in Prostate Cancer Cells Through the Mitochondria-Mediated Pathway. Cancer Biother Radiopharm 2014; 29:345-50. [PMID: 25379610 DOI: 10.1089/cbr.2014.1661] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Junying Zhou
- Department of Urology, People's Hospital of Zhengzhou City, Zhengzhou, China
| | - Fuhua Zhang
- Department of Urology, People's Hospital of Zhengzhou City, Zhengzhou, China
| | - Xiaoli Hou
- Department of Urology, People's Hospital of Zhengzhou City, Zhengzhou, China
| | - Nan Zhang
- Department of Urology, People's Hospital of Zhengzhou City, Zhengzhou, China
| |
Collapse
|
43
|
Jiang X, Zhong W, Huang H, He H, Jiang F, Chen Y, Yue F, Zou J, Li X, He Y, You P, Yang W, Lai Y, Wang F, Liu L. Autophagy defects suggested by low levels of autophagy activator MAP1S and high levels of autophagy inhibitor LRPPRC predict poor prognosis of prostate cancer patients. Mol Carcinog 2014; 54:1194-204. [PMID: 25043940 DOI: 10.1002/mc.22193] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 05/16/2014] [Accepted: 05/22/2014] [Indexed: 11/11/2022]
Abstract
MAP1S (originally named C19ORF5) is a widely distributed homolog of neuronal-specific MAP1A and MAP1B, and bridges autophagic components with microtubules and mitochondria to affect autophagosomal biogenesis and degradation. Mitochondrion-associated protein LRPPRC functions as an inhibitor for autophagy initiation to protect mitochondria from autophagy degradation. MAP1S and LRPPRC interact with each other and may collaboratively regulate autophagy although the underlying mechanism is yet unknown. Previously, we have reported that LRPPRC levels serve as a prognosis marker of patients with prostate adenocarcinomas (PCA), and that patients with high LRPPRC levels survive a shorter period after surgery than those with low levels of LRPPRC. MAP1S levels are elevated in diethylnitrosamine-induced hepatocelular carcinomas in wildtype mice and the exposed MAP1S-deficient mice develop more malignant hepatocellular carcinomas. We performed immunochemical analysis to evaluate the co-relationship among the levels of MAP1S, LRPPRC, P62, and γ-H2AX. Samples were collected from wildtype and prostate-specific PTEN-deficient mice, 111 patients with PCA who had been followed up for 10 years and 38 patients with benign prostate hyperplasia enrolled in hospitals in Guangzhou, China. The levels of MAP1S were generally elevated so the MAP1S-mediated autophagy was activated in PCA developed in either PTEN-deficient mice or patients than their respective benign tumors. The MAP1S levels among patients with PCA vary dramatically, and patients with low MAP1S levels survive a shorter period than those with high MAP1S levels. Levels of MAP1S in collaboration with levels of LRPPRC can serve as markers for prognosis of prostate cancer patients.
Collapse
Affiliation(s)
- Xianhan Jiang
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, China.,Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, 77030
| | - Weide Zhong
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, 510180, China
| | - Hai Huang
- Department of Urology, The Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Huichan He
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, 510180, China
| | - Funeng Jiang
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, 510180, China
| | - Yanru Chen
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, 510180, China
| | - Fei Yue
- Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, 77030
| | - Jing Zou
- Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, 77030
| | - Xun Li
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, China
| | - Yongzhong He
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, China
| | - Pan You
- Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, 77030
| | - Weiqiang Yang
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, China
| | - Yiming Lai
- Department of Urology, The Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Fen Wang
- Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, 77030
| | - Leyuan Liu
- Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, 77030
| |
Collapse
|
44
|
Zou J, Yue F, Li W, Song K, Jiang X, Yi J, Liu L. Autophagy inhibitor LRPPRC suppresses mitophagy through interaction with mitophagy initiator Parkin. PLoS One 2014; 9:e94903. [PMID: 24722279 PMCID: PMC3983268 DOI: 10.1371/journal.pone.0094903] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 03/21/2014] [Indexed: 12/19/2022] Open
Abstract
Autophagy plays an important role in tumorigenesis. Mitochondrion-associated protein LRPPRC interacts with MAP1S that interacts with LC3 and bridges autophagy components with microtubules and mitochondria to affect autophagy flux. Dysfunction of LRPPRC and MAP1S is associated with poor survival of ovarian cancer patients. Furthermore, elevated levels of LRPPRC predict shorter overall survival in patients with prostate adenocarcinomas or gastric cancer. To understand the role of LRPPRC in tumor development, previously we reported that LRPPRC forms a ternary complex with Beclin 1 and Bcl-2 to inhibit autophagy. Here we further show that LRPPRC maintains the stability of Parkin that mono-ubiquitinates Bcl-2 to increase Bcl-2 stability to inhibit autophagy. Under mitophagy stress, Parkin translocates to mitochondria to cause rupture of outer mitochondrial membrane and bind with exposed LRPPRC. Consequently, LRPPRC and Parkin help mitochondria being engulfed in autophagosomes to be degraded. In cells under long-term mitophagy stress, both LRPPRC and Parkin become depleted coincident with disappearance of mitochondria and final autophagy inactivation due to depletion of ATG5-ATG12 conjugates. LRPPRC functions as a checkpoint protein that prevents mitochondria from autophagy degradation and impact tumorigenesis.
Collapse
Affiliation(s)
- Jing Zou
- Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, United States of America
- Jiangxi Research Institute of Ophthalmology and Visual Sciences, The Affiliated Eye Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Fei Yue
- Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, United States of America
| | - Wenjiao Li
- Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, United States of America
| | - Kun Song
- Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, United States of America
| | - Xianhan Jiang
- Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, United States of America
| | - Jinglin Yi
- Jiangxi Research Institute of Ophthalmology and Visual Sciences, The Affiliated Eye Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Leyuan Liu
- Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|