1
|
Zhao Y, Guo M, Pei T, Shang C, Chen Y, Zhao L, Lu Y, Liang C, Wang J, Zhang J. α-Lipoic Acid Ameliorates Arsenic-Induced Lipid Disorders by Promoting Peroxisomal β-Oxidation and Reducing Lipophagy in Chicken Hepatocyte. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2413255. [PMID: 39887668 DOI: 10.1002/advs.202413255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 12/18/2024] [Indexed: 02/01/2025]
Abstract
Liver disease poses a significant threat to global public health, with arsenic (As) recognized as a major environmental toxin contributing to liver injury. However, the specific mechanisms and the protective effects of α-lipoic acid (LA) remain unclear. Therefore, this study employs network toxicology and network pharmacology to comprehensively analyze the hepatotoxic mechanism of As and the hepatoprotective mechanism of LA, and further verifies the mechanisms of peroxisomal β-oxidation and lipophagy in the process. The network analysis results show that As induces liver damage mainly through autophagy, apoptosis, lipid metabolism, and oxidative stress, whereas LA exerts its hepatoprotective properties mainly by regulating lipid metabolism. Further verifications find that As inhibits SIRT1 expression, activates the P53 and Notch pathways, damages mitochondria, inhibits peroxisomal β-oxidation, increases lipid accumulation, and enhances lipophagy in the liver, while LA intervention alleviates As-induced lipid accumulation and enhances lipophagy by targeting SIRT1, ameliorating mitochondrial damage, enhancing peroxisomal β-oxidation, thereby alleviating As-induced liver damage. This study further clarifies the mechanism of As hepatotoxicity and provides a theoretical basis for LA as a potential hepatoprotective agent.
Collapse
Affiliation(s)
- Yangfei Zhao
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Mingyue Guo
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Ting Pei
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Chenqi Shang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Yirong Chen
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Liying Zhao
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Yiguang Lu
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Chen Liang
- College of Animal Science, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Jundong Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Jianhai Zhang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| |
Collapse
|
2
|
Gao G, Miao J, Jia Y, He A. Mitochondria-associated programmed cell death: elucidating prognostic biomarkers, immune checkpoints, and therapeutic avenues in multiple myeloma. Front Immunol 2024; 15:1448764. [PMID: 39726602 PMCID: PMC11670199 DOI: 10.3389/fimmu.2024.1448764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
Background Multiple myeloma (MM) is a hematological malignancy characterized by the abnormal proliferation of plasma cells. Mitochondrial dysfunction and dysregulated programmed cell death (PCD) pathways have been implicated in MM pathogenesis. However, the precise roles of mitochondria-related genes (MRGs) and PCD-related genes (PCDRGs) in MM prognosis remain unclear. Methods Transcriptomic data from MM patients and healthy controls were analyzed to identify differentially expressed genes (DEGs). Candidate genes were selected by intersecting DEGs with curated lists of MRGs and PCDRGs. Univariate Cox, least absolute shrinkage and selection operator (LASSO), multivariate Cox, and stepwise regression analyses identified prognostic genes among the candidates. A risk model was constructed from these genes, and patients were stratified into high- and low-risk groups for survival analysis. Independent prognostic factors were incorporated into a nomogram to predict MM patient outcomes. Model performance was evaluated using calibration curves, receiver operating characteristic (ROC) analysis, and decision curve analysis (DCA). Finally, associations between prognostic genes and immune cell infiltration/drug responses were explored. Results 2,192 DEGs were detected between MM and control samples. 30 candidate genes were identified at the intersection of DEGs, 1,136 MRGs, and 1,548 PCDRGs. TRIAP1, TOMM7, PINK1, CHCHD10, PPIF, BCL2L1, and NDUFA13 were selected as prognostic genes. The risk model stratified patients into high- and low-risk groups with significantly different survival probabilities. Age, gender, ISS stage, and risk score were independent prognostic factors. The nomogram displayed good calibration and discriminative ability (AUC) in predicting survival, with clinical utility demonstrated by DCA. 9 immune cell types showed differential infiltration between MM and controls, with significant associations to risk scores and specific prognostic genes. 57 drugs, including nelarabine and vorinostat, were predicted to interact with the prognostic genes. Ultimately, qPCR in clinical samples from MM patients and healthy donors validated the expression levels of the seven key prognostic genes, corroborating the bioinformatic findings. Conclusion Seven genes (TRIAP1, TOMM7, PINK1, CHCHD10, PPIF, BCL2L1, NDUFA13) involved in mitochondrial function and PCD pathways were identified as prognostic markers in MM. These findings provide insights into MM biology and prognosis, highlighting potential therapeutic targets.
Collapse
Affiliation(s)
- Gongzhizi Gao
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jiyu Miao
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yachun Jia
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Aili He
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- National-Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Xi’an Key Laboratory of Hematological Diseases, Xi’an, China
| |
Collapse
|
3
|
Gao Y, Wei H, Peng X, Wang C, Zhu H, Yin J. ER stress-induced YAP upregulation leads to chondrocyte phenotype loss in age-related osteoarthritis. Front Pharmacol 2024; 15:1476255. [PMID: 39600372 PMCID: PMC11588467 DOI: 10.3389/fphar.2024.1476255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 10/25/2024] [Indexed: 11/29/2024] Open
Abstract
Background Osteoarthritis (OA) is a common degenerative joint disease, leading to pain and restricted mobility. Age-related endoplasmic reticulum (ER) stress has been implicated in the pathogenesis of OA, but the underlying mechanisms remain unclear. This study aims to explore the relationship between age-related ER stress, YAP overexpression, and chondrocyte phenotype loss in the development of OA. Methods Cartilage samples were collected from patients undergoing amputation, and age-related ER stress markers and YAP expression were assessed using immunohistochemical staining and qPCR. Transgenic mice with cartilage-specific YAP overexpression (YAPOE) were created, and Pamrevlumab was administered to evaluate its therapeutic effects. Results Higher expression of ER stress markers and YAP were showed in aged tissues compared to younger tissues. YAP overexpression led to decreased levels of cartilage phenotype markers and increased osteogenesis-related proteins. In vivo, YAPOE mice exhibited OA-like cartilage degeneration, which was mitigated by Pamrevlumab treatment. Conclusion Age-related ER stress induces YAP overexpression, contributing to OA pathogenesis. Pamrevlumab effectively prevents this phenotype loss in YAPOE mice, suggesting its potential as a therapeutic agent for OA. These findings provide new insights into the molecular mechanisms of OA and highlight the importance of targeting the ER stress-YAP-CTGF signaling pathway in OA treatment and prevention.
Collapse
Affiliation(s)
- Yanchun Gao
- Department of Orthopaedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haifeng Wei
- Department of Orthopaedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyuan Peng
- Department of Orthopaedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chenchen Wang
- Department of Orthopedics Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongyi Zhu
- Department of Orthopaedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junhui Yin
- Department of Orthopaedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
4
|
Wang Y, Xu J, Fu Z, Zhang R, Zhu W, Zhao Q, Wang P, Hu C, Cheng X. The role of reactive oxygen species in gastric cancer. Cancer Biol Med 2024; 21:j.issn.2095-3941.2024.0182. [PMID: 38982978 DOI: 10.20892/j.issn.2095-3941.2024.0182] [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] [Indexed: 07/11/2024] Open
Abstract
Gastric cancer (GC) ranks fifth in cancer incidence and fourth in cancer-related mortality worldwide. Reactive oxygen species (ROS) are highly oxidative oxygen-derived products that have crucial roles in cell signaling regulation and maintaining internal balance. ROS are closely associated with the occurrence, development, and treatment of GC. This review summarizes recent findings on the sources of ROS and the bidirectional regulatory effects on GC and discusses various treatment modalities for GC that are related to ROS induction. In addition, the regulation of ROS by natural small molecule compounds with the highest potential for development and applications in anti-GC research is summarized. The aim of the review is to accelerate the clinical application of modulating ROS levels as a therapeutic strategy for GC.
Collapse
Affiliation(s)
- Yuqi Wang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou 310022, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Jingli Xu
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou 310022, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Zhenjie Fu
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou 310022, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Ruolan Zhang
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou 310022, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Weiwei Zhu
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou 310022, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Qianyu Zhao
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou 310022, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Ping Wang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Can Hu
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou 310022, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Xiangdong Cheng
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou 310022, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| |
Collapse
|
5
|
Gao G, Liu R, Wu D, Gao D, Lv Y, Xu X, Fu B, Lin Z, Wang T, He A, Bai J. Risk score constructed with neutrophil extracellular traps-related genes predicts prognosis and immune microenvironment in multiple myeloma. Front Oncol 2024; 14:1365460. [PMID: 38919521 PMCID: PMC11196624 DOI: 10.3389/fonc.2024.1365460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 05/28/2024] [Indexed: 06/27/2024] Open
Abstract
Background Multiple myeloma (MM) exhibits considerable heterogeneity in treatment responses and survival rates, even when standardized care is administered. Ongoing efforts are focused on developing prognostic models to predict these outcomes more accurately. Recently, neutrophil extracellular traps (NETs) have emerged as a potential factor in MM progression, sparking investigation into their role in prognostication. Methods In this study, a multi-gene risk scoring model was constructed using the intersection of NTEs and differentially expressed genes (DEGs), applying the least absolute shrinkage and selection operator (LASSO) Cox regression model. A nomogram was established, and the prognostic model's effectiveness was determined via Kaplan-Meier survival analysis, receiver operating characteristic (ROC) curve, and decision curve analysis (DCA). The ESTIMATE algorithm and immune-related single-sample gene set enrichment analysis (ssGSEA) were employed to evaluate the level of immune infiltration. The sensitivity of chemotherapy drugs was assessed using the Genomics of Drug Sensitivity in Cancer (GDSC) database. Ultimately, the presence of the detected genes was confirmed through quantitative real-time polymerase chain reaction (qRT-PCR) analysis in MM cell specimens. Results 64 NETs-DEGs were yielded, and through univariate Cox regression and LASSO regression analysis, we constructed a risk score composed of six genes: CTSG, HSPE1, LDHA, MPO, PINK1, and VCAM1. MM patients in three independent datasets were classified into high- and low-risk groups according to the risk score. The overall survival (OS) of patients in the high-risk group was significantly reduced compared to the low-risk group. Furthermore, the risk score was an independent predictive factor for OS. In addition, interactions between the risk score, immune score, and immune cell infiltration were investigated. Further analysis indicated that patients in the high-risk group were more sensitive to a variety of chemotherapy and targeted drugs, including bortezomib. Moreover, the six genes provided insights into the progression of plasma cell disorders. Conclusion This study offers novel insights into the roles of NETs in prognostic prediction, immune status, and drug sensitivity in MM, serving as a valuable supplement and enhancement to existing grading systems.
Collapse
Affiliation(s)
- Gongzhizi Gao
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Rui Liu
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Dong Wu
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Dandan Gao
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yang Lv
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xuezhu Xu
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Bingjie Fu
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Zujie Lin
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Ting Wang
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Aili He
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- National-Local Joint Engineering Research Center of Biodiagnostics & Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Xi’an Key Laboratory of hematological diseases, Xi’an, China
| | - Ju Bai
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Xi’an Key Laboratory of hematological diseases, Xi’an, China
| |
Collapse
|
6
|
Li W, Huang C, Qiu L, Tang Y, Zhang X, Zhang L, Zhao H, Miyagishi M, Kasim V, Wu S. p52-ZER6/IGF1R axis maintains cancer stem cell population to promote cancer progression by enhancing pro-survival mitophagy. Oncogene 2024; 43:2115-2131. [PMID: 38773262 DOI: 10.1038/s41388-024-03058-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 04/27/2024] [Accepted: 05/01/2024] [Indexed: 05/23/2024]
Abstract
Cancer stem cells (CSCs), which are distinct subpopulations of tumor cells, have a substantially higher tumor-initiating capacity and are closely related to poor clinical outcomes. Damage to organelles can trigger CSC pool exhaustion; however, the underlying mechanisms are poorly understood. ZER6 is a zinc-finger protein with two isoforms possessing different amino termini: p52-ZER6 and p71-ZER6. Since their discovery, almost no study reported on their biological and pathological functions. Herein, we found that p52-ZER6 was crucial for CSC population maintenance; p52-ZER6-knocking down almost abolished the tumor initiation capability. Through transcriptomic analyses together with in vitro and in vivo studies, we identified insulin like growth factor 1 receptor (IGF1R) as the transcriptional target of p52-ZER6 that mediated p52-ZER6 regulation of CSC by promoting pro-survival mitophagy. Moreover, this regulation of mitophagy-mediated CSC population maintenance is specific to p52-ZER6, as p71-ZER6 failed to exert the same effect, most possibly due to the presence of the HUB1 domain at its N-terminus. These results provide a new perspective on the regulatory pathway of pro-survival mitophagy in tumor cells and the molecular mechanism underlying p52-ZER6 oncogenic activity, suggesting that targeting p52-ZER6/IGF1R axis to induce CSC pool exhaustion may be a promising anti-tumor therapeutic strategy.
Collapse
Affiliation(s)
- Wenfang Li
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
- School of Pharmaceutical Sciences and Institute of Materia Medica, Xinjiang University, Urumqi, 830017, China
| | - Can Huang
- Metabolic Disease Research Center, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, 230032, China.
| | - Li Qiu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Yu Tang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Xia Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Lei Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Hezhao Zhao
- Department of Gastrointestinal Surgery, Chongqing University Cancer Hospital, Chongqing University, Chongqing, 400030, China
| | - Makoto Miyagishi
- Life Science Innovation, School of Integrative and Global Majors, University of Tsukuba, Tsukuba, Ibaraki, 305-0006, Japan
| | - Vivi Kasim
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing University, Chongqing, 400030, China.
| | - Shourong Wu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing University, Chongqing, 400030, China.
| |
Collapse
|
7
|
Zhao MM, Wang B, Huang WX, Zhang L, Peng R, Wang C. Verteporfin suppressed mitophagy via PINK1/parkin pathway in endometrial cancer. Am J Cancer Res 2024; 14:1935-1946. [PMID: 38726279 PMCID: PMC11076261 DOI: 10.62347/pmyv3832] [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: 02/04/2024] [Accepted: 03/15/2024] [Indexed: 05/12/2024] Open
Abstract
Endometrial cancer (EC) is a malignancy that poses a threat to woman's health worldwide. Building upon prior work, we explored the inhibitory effect of verteporfin on EC. We showed that verteporfin can damage the mitochondria of EC cells, leading to a decrease of mitochondrial membrane potential and an increase in ROS (reactive oxygen species). In addition, verteporfin treatment was shown to inhibit the proliferation and migration of EC cells, promote apoptosis, and reduce the expression of mitophagy-related proteins PINK1/parkin and TOM20. The ROS inhibitor N-Acetyl Cysteine was able to rescue the expression of PINK1/parkin proteins. This suggests that verteporfin may inhibit mitophagy by elevating ROS levels, thereby inhibiting EC cell viability. The effect of verteporfin on mitophagy supports further investigation as a potential therapeutic option for EC.
Collapse
Affiliation(s)
- Ming-Ming Zhao
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan UniversityShanghai 200011, China
| | - Bo Wang
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan UniversityShanghai 200011, China
| | - Wen-Xi Huang
- School of Basic Medical Sciences, Shanghai Medical College, Fudan UniversityShanghai 200032, China
| | - Li Zhang
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan UniversityShanghai 200011, China
| | - Rui Peng
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan UniversityShanghai 200011, China
| | - Chao Wang
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan UniversityShanghai 200011, China
| |
Collapse
|
8
|
Booth L, Roberts JL, West C, Dent P. GZ17-6.02 interacts with proteasome inhibitors to kill multiple myeloma cells. Oncotarget 2024; 15:159-174. [PMID: 38441437 PMCID: PMC10913917 DOI: 10.18632/oncotarget.28558] [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: 12/01/2023] [Accepted: 01/23/2024] [Indexed: 03/07/2024] Open
Abstract
GZ17-6.02, a synthetically manufactured compound containing isovanillin, harmine and curcumin, has undergone phase I evaluation in patients with solid tumors (NCT03775525) with a recommended phase 2 dose (RP2D) of 375 mg PO BID. GZ17-6.02 was more efficacious as a single agent at killing multiple myeloma cells than had previously been observed in solid tumor cell types. GZ17-6.02 interacted with proteasome inhibitors in a greater than additive fashion to kill myeloma cells and alone it killed inhibitor-resistant cells to a similar extent. The drug combination of GZ17-6.02 and bortezomib activated ATM, the AMPK and PERK and inactivated ULK1, mTORC1, eIF2α, NFκB and the Hippo pathway. The combination increased ATG13 S318 phosphorylation and the expression of Beclin1, ATG5, BAK and BIM, and reduced the levels of BCL-XL and MCL1. GZ17-6.02 interacted with bortezomib to enhance autophagosome formation and autophagic flux, and knock down of ATM, AMPKα, ULK1, Beclin1 or ATG5 significantly reduced both autophagy and tumor cell killing. Knock down of BAK and BIM significantly reduced tumor cell killing. The expression of HDACs1/2/3 was significantly reduced beyond that previously observed in solid tumor cells and required autophagy. This was associated with increased acetylation and methylation of histone H3. Combined knock down of HDACs1/2/3 caused activation of ATM and the AMPK and caused inactivation of ULK1, mTORC1, NFκB and the Hippo pathway. HDAC knock down also enhanced ATG13 phosphorylation, increased BAK levels and reduced those of BCL-XL. Collectively, our present studies support performing additional in vivo studies with multiple myeloma cells.
Collapse
Affiliation(s)
- Laurence Booth
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Jane L. Roberts
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Cameron West
- Genzada Pharmaceuticals, Hutchinson, KS 67502, USA
| | - Paul Dent
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
9
|
He Z, Shi J, Zhu B, Tian Z, Zhang Z. Transcriptomics analysis revealed that TAZ regulates the proliferation of KIRC cells through mitophagy. BMC Cancer 2024; 24:229. [PMID: 38373978 PMCID: PMC10875871 DOI: 10.1186/s12885-024-11903-9] [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: 08/09/2023] [Accepted: 01/20/2024] [Indexed: 02/21/2024] Open
Abstract
Transcriptional Co-Activator with PDZ-Binding Motif (TAZ, also known as WWTR1) is a downstream effector of the Hippo pathway, involved in the regulation of organ regeneration and cell differentiation in processes such as development and regeneration. TAZ has been shown to play a tumor-promoting role in various cancers. Currently, many studies focus on the role of TAZ in the process of mitophagy. However, the molecular mechanism and biological function of TAZ in renal clear cell carcinoma (KIRC) are still unclear. Therefore, we systematically analyzed the mRNA expression profile and clinical data of KIRC in The Cancer Genome Atlas (TCGA) dataset. We found that TAZ expression was significantly upregulated in KIRC compared with normal kidney tissue and was closely associated with poor prognosis of patients. Combined with the joint analysis of 36 mitophagy genes, it was found that TAZ was significantly negatively correlated with the positive regulators of mitophagy. Finally, our results confirmed that high expression of TAZ in KIRC inhibits mitophagy and promotes KIRC cell proliferation. In conclusion, our findings reveal the important role of TAZ in KIRC and have the potential to be a new target for KIRC therapy.
Collapse
Affiliation(s)
- Zhen He
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
- Department of Urology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Jianxi Shi
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Bing Zhu
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zhentao Tian
- Department of Urology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Zhihong Zhang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China.
| |
Collapse
|
10
|
Lv T, Zhang H. Mitophagy-related gene signature for predicting the prognosis of multiple myeloma. Heliyon 2024; 10:e24520. [PMID: 38317923 PMCID: PMC10838706 DOI: 10.1016/j.heliyon.2024.e24520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 11/26/2023] [Accepted: 01/10/2024] [Indexed: 02/07/2024] Open
Abstract
Aims The aims of this study were to explore the molecular mechanism of mitophagy in multiple myeloma (MM) and to develop an effective prognostic signature for the disease based on mitophagy-related genes (MRGs). Methods Three gene sets from the Reactome database were used to explore MRGs, following which those that were differentially expressed between MM and normal samples were investigated using the data from the Genomic Data Commons-Multiple Myeloma Research Foundation-CoMMpass Study. Mitophagy-related molecular subtypes of MM were identified and their immune infiltration, associated patient survival rates, immune checkpoint genes, and mitophagy scores were compared. Prognostic genes for MM were identified, and a prognostic model was constructed. Additionally, a nomogram was constructed using the prognostic model and prognosis-related clinical features. Finally, the drug sensitivity and correlation analyses of the subtypes were performed between the two risk groups. Results We identified two MM molecular subtypes that exhibited significant differences in mitophagy scores, associated patient survival rates, immune infiltration, and immune checkpoint genes. An MRG-based prognostic signature was constructed using six genes (TRIP13, KIF7, GPR63, CRIP2, DNTT, and HSPB8), which had high predictive prognostic value. A nomogram was constructed by screening five indicators (risk score, subtype, age, sex, and stage) that could predict the 1-, 3-, and 5-year survival probabilities of patients with MM. The two risk groups displayed significant differences in their IC50 values of 33 drugs, such as bleomycin. Patients in the high-risk group tended to fall within Mitophagy_cluster_A. Conclusion Our MRG-based signature is a promising prognostic biomarker for MM.
Collapse
Affiliation(s)
- Tiange Lv
- Cadre's Ward, The General Hospital of Northern Theater Command, Shenyang, Liaoning, 110015, China
| | - Haocong Zhang
- Department of Orthopaedics, The General Hospital of Northern Theater Command, Shenyang, Liaoning, 110015, China
| |
Collapse
|
11
|
Yang HB, Li Y, Li XH, Yan QM, Han XZ, Cao J, Sang HP, Li JL. The compensatory increase of Gli-similar 3 inhibited neuronal apoptosis through regulating Mps one binder kinase activator 1b (MOB1b): a possible strategy for the functional recovery after spinal cord injury. Exp Anim 2024; 73:61-72. [PMID: 37574269 PMCID: PMC10877155 DOI: 10.1538/expanim.23-0041] [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: 03/16/2023] [Accepted: 08/02/2023] [Indexed: 08/15/2023] Open
Abstract
Spinal cord injury (SCI) is a devastating disease characterized by neuronal apoptosis. Gli-similar 3 (GLIS3), a transcriptional factor, was involved in cell apoptosis and associated with the transcription of downstream target genes related to neuronal function. However, the function of GLIS3 in SCI remains unknown. Therefore, we used the mouse model of SCI to explore the role of GLIS3 in SCI. The results showed that GLIS3 expression was significantly increased in spinal cord tissues of SCI mice, and GLIS3 overexpression promoted the functional recovery, reserved histological changes, and inhibited neuronal apoptosis after SCI. Through online tools, the potential target genes of GLIS3 were analyzed and we found that Mps one binder kinase activator 1b (Mob1b) had a strong association with SCI among these genes. MOB1b is a core component of Hippo signaling pathway, which was reported to inhibit cell apoptosis. MOB1b expression was significantly increased in mice at 7 days post-SCI and GLIS3 overexpression further increased its expression. Dual-luciferase reporter assay revealed that GLIS3 bound to the promoter of Mob1b and promoted its transcription. In conclusion, our findings reveal that the compensatory increase of GLIS3 promotes functional recovery after SCI through inhibiting neuronal apoptosis by transcriptionally regulating MOB1b. Our study provides a novel target for functional recovery after SCI.
Collapse
Affiliation(s)
- Hong-Bo Yang
- Department of Orthopedics, Affiliated Hospital of Chifeng University, No. 42 Wangfu Street, Songshan District, Chifeng, Inner Mongolia, 024000, P.R. China
- Institute of Orthopedic Diseases, Chifeng University, No.1 Yingbin Road, Hongshan District, Chifeng, 024000, P.R. China
| | - Ying Li
- Department of Neurology, Affiliated Hospital of Chifeng University, No. 42 Wangfu Street, Songshan District, Chifeng, 024000, P.R. China
| | - Xiu-Hai Li
- Department of Orthopedics, Affiliated Hospital of Chifeng University, No. 42 Wangfu Street, Songshan District, Chifeng, Inner Mongolia, 024000, P.R. China
| | - Qing-Ming Yan
- Department of Orthopedics, Affiliated Hospital of Chifeng University, No. 42 Wangfu Street, Songshan District, Chifeng, Inner Mongolia, 024000, P.R. China
| | - Xian-Zhang Han
- Department of Orthopedics, Affiliated Hospital of Chifeng University, No. 42 Wangfu Street, Songshan District, Chifeng, Inner Mongolia, 024000, P.R. China
| | - Jian Cao
- Department of Orthopedics, Affiliated Hospital of Chifeng University, No. 42 Wangfu Street, Songshan District, Chifeng, Inner Mongolia, 024000, P.R. China
- Institute of Orthopedic Diseases, Chifeng University, No.1 Yingbin Road, Hongshan District, Chifeng, 024000, P.R. China
| | - Hong-Peng Sang
- Department of Orthopedics, Affiliated Hospital of Chifeng University, No. 42 Wangfu Street, Songshan District, Chifeng, Inner Mongolia, 024000, P.R. China
- Institute of Orthopedic Diseases, Chifeng University, No.1 Yingbin Road, Hongshan District, Chifeng, 024000, P.R. China
| | - Jin-Long Li
- Department of Orthopedics, Affiliated Hospital of Chifeng University, No. 42 Wangfu Street, Songshan District, Chifeng, Inner Mongolia, 024000, P.R. China
- Institute of Orthopedic Diseases, Chifeng University, No.1 Yingbin Road, Hongshan District, Chifeng, 024000, P.R. China
| |
Collapse
|
12
|
Zhang F, Issah MA, Fu HY, Zhou HR, Liu TB, Shen JZ. LATS1 Promotes B-ALL Tumorigenesis by Regulating YAP1 Phosphorylation and Subcellular Localization. Curr Med Sci 2024; 44:81-92. [PMID: 38277019 DOI: 10.1007/s11596-023-2821-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 11/15/2023] [Indexed: 01/27/2024]
Abstract
OBJECTIVE YAP1 plays a dual role as an oncogene and tumor suppressor gene in several tumors; differentiating between these roles may depend on the YAP1 phosphorylation pattern. The specific function of YAP1 in B cell acute lymphoblastic leukemia (B-ALL), however, is currently unclear. Thus, in the present study, the role of YAP1 in B-ALL was investigated using relevant cell lines and patient datasets. METHODS The effects of shRNA-mediated knockdown on YAP1 and LATS1 levels in the NALM6 and MOLT-4 cell lines were examined using Western blotting, quantitative real-time polymerase chain reaction, flow cytometry, immunostaining, and nude mouse subcutaneous tumorigenesis experiments. Gene expression levels of Hippo pathway-related molecules before and after verteporfin (VP) treatment were compared using RNA-Seq to identify significant Hippo pathway-related genes in NALM6 cells. RESULTS Patients with ALL showing high YAP1 expression and low YAP1-Ser127 phosphorylation levels had worse prognoses than those with low YAP1 protein expression and high YAP1-Ser127 phosphorylation levels. YAP1-Ser127 phosphorylation levels were lower in NALM6 cells than in MOLT-4 and control cells; YAP1 was distributed in the nuclei in NALM6 cells. Knockdown of YAP1 inhibited MOLT-4 and NALM6 cell proliferation and arrested the NALM6 cell cycle in the G0/G1 phase. Before and after VP treatment, the expression of the upstream gene LATS1 was upregulated; its overexpression promoted YAP1-Ser127 phosphorylation. Further, YAP1 was distributed in the plasma. CONCLUSION LATS1 may downregulate YAP1-Ser127 phosphorylation and maintain B-ALL cell function; thus, VP, which targets this axis, may serve as a new therapeutic method for improving the outcomes for B-ALL patients.
Collapse
Affiliation(s)
- Feng Zhang
- Fujian Provincial Key Laboratory on Hematology, Fujian Medical Center of Hematology, Fujian Institute of Hematology, Clinical Research Center for Hematological Malignancies of Fujian Province, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Mohammed Awal Issah
- Tamale Technical University, Faculty of Allied Health and Pharmaceutical Sciences, Department of Medical Laboratory Technology, Tamale, NS-011-2000, Ghana
| | - Hai-Ying Fu
- Department of Hematology, The Third Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, The Third People's Hospital of Fujian Province, Fuzhou, 350122, China
| | - Hua-Rong Zhou
- Fujian Provincial Key Laboratory on Hematology, Fujian Medical Center of Hematology, Fujian Institute of Hematology, Clinical Research Center for Hematological Malignancies of Fujian Province, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Ting-Bo Liu
- Fujian Provincial Key Laboratory on Hematology, Fujian Medical Center of Hematology, Fujian Institute of Hematology, Clinical Research Center for Hematological Malignancies of Fujian Province, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Jian-Zhen Shen
- Fujian Provincial Key Laboratory on Hematology, Fujian Medical Center of Hematology, Fujian Institute of Hematology, Clinical Research Center for Hematological Malignancies of Fujian Province, Fujian Medical University Union Hospital, Fuzhou, 350001, China.
| |
Collapse
|
13
|
Tan XL, Wang Z, Liao S, Yi M, Tao D, Zhang X, Leng X, Shi J, Xie S, Yang Y, Liu YQ. NR0B1 augments sorafenib resistance in hepatocellular carcinoma through promoting autophagy and inhibiting apoptosis. Cancer Sci 2024; 115:465-476. [PMID: 37991109 PMCID: PMC10859617 DOI: 10.1111/cas.16029] [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: 06/21/2023] [Revised: 11/06/2023] [Accepted: 11/10/2023] [Indexed: 11/23/2023] Open
Abstract
NR0B1 is frequently activated in hepatocellular carcinoma (HCC). However, the role of NR0B1 is controversial in HCC. In this study, we observed that NR0B1 was an independent poor prognostic factor, negatively correlated with the overall survival of HCC and the relapse-free survival of patients treated with sorafenib. Meanwhile, NR0B1 promoted the proliferation, migration, and invasion of HCC cells, inhibited sorafenib-induced apoptosis, and elevated the IC50 of sorafenib in HCC cells. NR0B1 was further displayed to increase sorafenib-induced autophagic vesicles and activate Beclin1/LC3-II-dependent autophagy pathway. Finally, NR0B1 was revealed to transcriptionally suppress GSK3β that restrains AMPK/mTOR-driven autophagy and increases BAX-mediated apoptosis. Collectively, our study uncovered that the ectopic expression of NR0B1 augmented sorafenib-resistance in HCC cells by activating autophagy and inhibiting apoptosis. Our findings supported that NR0B1 was a detrimental factor for HCC prognosis.
Collapse
Affiliation(s)
- Xiao lan Tan
- Department of Medical Genetics and State Key Laboratory of Biotherapy, West China HospitalSichuan UniversityChengduChina
| | - Zhaokun Wang
- Department of Medical Genetics and State Key Laboratory of Biotherapy, West China HospitalSichuan UniversityChengduChina
| | - Shunyao Liao
- Institute of Gerontology and Center for Geriatrics, Sichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Ming Yi
- Department of Medical Genetics and State Key Laboratory of Biotherapy, West China HospitalSichuan UniversityChengduChina
| | - Dachang Tao
- Department of Medical Genetics and State Key Laboratory of Biotherapy, West China HospitalSichuan UniversityChengduChina
| | - Xinyue Zhang
- Department of Medical Genetics and State Key Laboratory of Biotherapy, West China HospitalSichuan UniversityChengduChina
| | - Xiangyou Leng
- Department of Medical Genetics and State Key Laboratory of Biotherapy, West China HospitalSichuan UniversityChengduChina
| | - Jiaying Shi
- Department of Medical Genetics and State Key Laboratory of Biotherapy, West China HospitalSichuan UniversityChengduChina
| | - Shengyu Xie
- Department of Medical Genetics and State Key Laboratory of Biotherapy, West China HospitalSichuan UniversityChengduChina
| | - Yuan Yang
- Department of Medical Genetics and State Key Laboratory of Biotherapy, West China HospitalSichuan UniversityChengduChina
| | - Yun qiang Liu
- Department of Medical Genetics and State Key Laboratory of Biotherapy, West China HospitalSichuan UniversityChengduChina
| |
Collapse
|
14
|
Wu C, Liu M, Liu J, Jia M, Zeng X, Fu Z, Geng Y, He Z, Zhang X, Yan H. Integrative analysis of an endoplasmic reticulum stress-related signature in multiple myeloma. J Gene Med 2024; 26:e3595. [PMID: 37730959 DOI: 10.1002/jgm.3595] [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: 07/23/2023] [Revised: 08/17/2023] [Accepted: 09/03/2023] [Indexed: 09/22/2023] Open
Abstract
BACKGROUND Multiple myeloma (MM) is a malignancy in which plasma cells proliferate abnormally, and it remains incurable. The cells are characterized by high levels of endoplasmic reticulum stress (ERS) and depend on the ERS response for survival. Thus, we aim to find an ERS-related signature of MM and assess its diagnostic value. METHODS We downloaded three datasets of MM from the Gene Expression Omnibus database. After identifying ERS-related differentially expressed genes (ERDEGs), we analyzed them using Gene Ontology enrichment analysis. A protein-protein interaction network, a transcription factor-mRNA network, a miRNA-mRNA network and a drug-mRNA network were constructed to explore the ERDEGs. The clinical application of these genes was identified by calculating the infiltration of immune cells and using receiver operating characteistic analyses. Finally, qPCR was performed to further confirm the roles of ERDEGs. RESULTS We obtained nine ERDEGs of MM. Gene Ontology enrichment indicated that the ERDEGs played a role in the endoplasmic reticulum membrane. Additionally, the protein-protein interaction network showed interaction among the ERDEGs, and there were 20 proteins, 107 transcription factors, 42 drugs or molecular compounds and 51 miRNAs which were likely to interact with the nine genes. In addition, immune cell infiltration analyses showed that there was a strong correlation between the nine genes and immune cells, and these potential biomarkers exhibited good diagnostic values. Finally, the expression of ERDEGs in MM cells was different from that in healthy donor samples. CONCLUSION The nine ERS-related genes, CR2, DHCR7, DNAJC3, KDELR2, LPL, OSBPL3, PINK1, VCAM1 and XBP1 are potential biomarkers of MM, and this supports further clinical development of the diagnosis and treatment of MM.
Collapse
Affiliation(s)
- Chengyu Wu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mei Liu
- Department of General Practice, Wuxi Branch of Ruijin Hospital, Jiangsu, China
| | - Jia Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingyuan Jia
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinyi Zeng
- Department of Hematology, Huadong Hospital Affiliated with Fudan University, Shanghai, China
| | - Ze Fu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanlai Geng
- Department of General Practice, Wuxi Branch of Ruijin Hospital, Jiangsu, China
| | - Ziqi He
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xian Zhang
- Department of General Practice, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hua Yan
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of General Practice, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
15
|
Jin S, Lee CJ, Lim G, Park S, Lee SH, Chung JH, Oh J, Kang SM. C-reactive protein accelerates DRP1-mediated mitochondrial fission by modulating ERK1/2-YAP signaling in cardiomyocytes. BMB Rep 2023; 56:663-668. [PMID: 37817437 PMCID: PMC10761750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/13/2023] [Accepted: 09/13/2023] [Indexed: 10/12/2023] Open
Abstract
C-reactive protein (CRP) is an inflammatory marker and risk factor for atherosclerosis and cardiovascular diseases. However, the mechanism through which CRP induces myocardial damage remains unclear. This study aimed to determine how CRP damages cardiomyocytes via the change of mitochondrial dynamics and whether survivin, an anti-apoptotic protein, exerts a cardioprotective effect in this process. We treated H9c2 cardiomyocytes with CRP and found increased intracellular ROS production and shortened mitochondrial length. CRP treatment phosphorylated ERK1/2 and promoted increased expression, phosphorylation, and translocation of DRP1, a mitochondrial fission-related protein, from the cytoplasm to the mitochondria. The expression of mitophagy proteins PINK1 and PARK2 was also increased by CRP. YAP, a transcriptional regulator of PINK1 and PARK2, was also increased by CRP. Knockdown of YAP prevented CRP-induced increases in DRP1, PINK1, and PARK2. Furthermore, CRP-induced changes in the expression of DRP1 and increases in YAP, PINK1, and PARK2 were inhibited by ERK1/2 inhibition, suggesting that ERK1/2 signaling is involved in CRP-induced mitochondrial fission. We treated H9c2 cardiomyocytes with a recombinant TAT-survivin protein before CRP treatment, which reduced CRP-induced ROS accumulation and reduced mitochondrial fission. CRP-induced activation of ERK1/2 and increases in the expression and activity of YAP and its downstream mitochondrial proteins were inhibited by TAT-survivin. This study shows that mitochondrial fission occurs during CRPinduced cardiomyocyte damage and that the ERK1/2-YAP axis is involved in this process, and identifies that survivin alters these mechanisms to prevent CRP-induced mitochondrial damage. [BMB Reports 2023; 56(12): 663-668].
Collapse
Affiliation(s)
- Suyeon Jin
- Division of Cardiology, Department of Internal Medicine, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Korea
- Graduate Program in Biomedical Engineering, Yonsei University College of Medicine, Seoul 03772, Korea
| | - Chan Joo Lee
- Division of Cardiology, Department of Internal Medicine, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Gibbeum Lim
- Division of Cardiology, Department of Internal Medicine, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Sungha Park
- Division of Cardiology, Department of Internal Medicine, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Sang-Hak Lee
- Division of Cardiology, Department of Internal Medicine, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Ji Hyung Chung
- Department of Applied Bioscience, College of Life Science, CHA University, Pocheon 11160, Korea
| | - Jaewon Oh
- Division of Cardiology, Department of Internal Medicine, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Seok-Min Kang
- Division of Cardiology, Department of Internal Medicine, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|
16
|
Zheng B, Yi K, Zhang Y, Pang T, Zhou J, He J, Lan H, Xian H, Li R. Multi-omics analysis of multiple myeloma patients with differential response to first-line treatment. Clin Exp Med 2023; 23:3833-3846. [PMID: 37515690 DOI: 10.1007/s10238-023-01148-4] [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: 05/29/2023] [Accepted: 07/20/2023] [Indexed: 07/31/2023]
Abstract
The genome backgrounds of multiple myeloma (MM) would affect the efficacy of specific treatment. However, the mutational and transcriptional landscapes in MM patients with differential response to first-line treatment remains unclear. We collected paired whole-exome sequencing (WES) and transcriptomic data of over 200 MM cases from MMRF-COMPASS project. R package, maftools was applied to analyze the somatic mutations and mutational signatures across MM samples. Differential expressed genes (DEG) was calculated using R package, DESeq2. The feature selection of the predictive model was determined by LASSO regression. In silico analysis revealed newly discovered recurrent mutated genes such as TTN, MUC16. TP53 mutation was observed more frequent in nonCR (complete remission) group with poor prognosis. DNA repair-associated mutational signatures were enriched in CR patients. Transcriptomic profiling showed that the activity of NF-kappa B and TGF-β pathways was suppressed in CR patients. A transcriptome-based response predictive model was constructed and showed promising predictive accuracy in MM patients receiving first-line treatment. Our study delineated distinctive mutational and transcriptional landscapes in MM patients with differential response to first-line treatment. Furthermore, we constructed a 20-gene predictive model which showed promising accuracy in predicting treatment response in newly diagnosed MM patients.
Collapse
Affiliation(s)
- Bo Zheng
- Nuclear Radiation Injury Protection and Treatment Department, Navy Medical Center of PLA, Naval Medical University, Huaihai West Road No. 338, Shanghai, 200050, China.
| | - Ke Yi
- Nuclear Radiation Injury Protection and Treatment Department, Navy Medical Center of PLA, Naval Medical University, Huaihai West Road No. 338, Shanghai, 200050, China
| | - Yajun Zhang
- Nuclear Radiation Injury Protection and Treatment Department, Navy Medical Center of PLA, Naval Medical University, Huaihai West Road No. 338, Shanghai, 200050, China
| | - Tongfang Pang
- Nuclear Radiation Injury Protection and Treatment Department, Navy Medical Center of PLA, Naval Medical University, Huaihai West Road No. 338, Shanghai, 200050, China
| | - Jieyi Zhou
- Nuclear Radiation Injury Protection and Treatment Department, Navy Medical Center of PLA, Naval Medical University, Huaihai West Road No. 338, Shanghai, 200050, China
| | - Jie He
- Nuclear Radiation Injury Protection and Treatment Department, Navy Medical Center of PLA, Naval Medical University, Huaihai West Road No. 338, Shanghai, 200050, China
| | - Hongyan Lan
- Nuclear Radiation Injury Protection and Treatment Department, Navy Medical Center of PLA, Naval Medical University, Huaihai West Road No. 338, Shanghai, 200050, China
| | - Hongming Xian
- Nuclear Radiation Injury Protection and Treatment Department, Navy Medical Center of PLA, Naval Medical University, Huaihai West Road No. 338, Shanghai, 200050, China
| | - Rong Li
- Nuclear Radiation Injury Protection and Treatment Department, Navy Medical Center of PLA, Naval Medical University, Huaihai West Road No. 338, Shanghai, 200050, China.
| |
Collapse
|
17
|
Jia Y, Liu R, Shi L, Feng Y, Zhang L, Guo N, He A, Kong G. Integrative analysis of the prognostic value and immune microenvironment of mitophagy-related signature for multiple myeloma. BMC Cancer 2023; 23:859. [PMID: 37700273 PMCID: PMC10496355 DOI: 10.1186/s12885-023-11371-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 09/04/2023] [Indexed: 09/14/2023] Open
Abstract
BACKGROUND Multiple myeloma (MM) is a fatal malignant tumor in hematology. Mitophagy plays vital roles in the pathogenesis and drug sensitivity of MM. METHODS We acquired transcriptomic expression data and clinical index of MM patients from NCI public database, and 36 genes involved in mitophagy from the gene set enrichment analysis (GSEA) database. Least absolute shrinkage and selection operator (LASSO) Cox regression analysis was conducted to construct a risk score prognostic model. Kaplan-Meier survival analysis and receiver operation characteristic curves (ROC) were conducted to identify the efficiency of prognosis and diagnosis. ESTIMATE algorithm and immune-related single-sample gene set enrichment analysis (ssGSEA) was performed to uncover the level of immune infiltration. QRT-PCR was performed to verify gene expression in clinical samples of MM patients. The sensitivity to chemotherapy drugs was evaluated upon the database of the genomics of drug sensitivity in cancer (GDSC). RESULTS Fifty mitophagy-related genes were differently expressed in two independent cohorts. Ten out of these genes were identified to be related to MM overall survival (OS) rate. A prognostic risk signature model was built upon on these genes: VDAC1, PINK1, VPS13C, ATG13, and HUWE1, which predicted the survival of MM accurately and stably both in training and validation cohorts. MM patients suffered more adverse prognosis showed more higher risk core. In addition, the risk score was considered as an independent prognostic element for OS of MM patients by multivariate cox regression analysis. Functional pathway enrichment analysis of differentially expressed genes (DEGs) based on risk score showed terms of cell cycle, immune response, mTOR pathway, and MYC targets were obviously enriched. Furthermore, MM patients with higher risk score were observed lower immune scores and lower immune infiltration levels. The results of qRT-PCR verified VDAC1, PINK1, and HUWE1 were dysregulated in new diagnosed MM patients. Finally, further analysis indicated MM patients showed more susceptive to bortezomib, lenalidomide and rapamycin in high-risk group. CONCLUSION Our research provided a neoteric prognostic model of MM based on mitophagy genes. The immune infiltration level based on risk score paved a better understanding of the participation of mitophagy in MM.
Collapse
Affiliation(s)
- Yachun Jia
- National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Rui Liu
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Luyi Shi
- Precision Medical Institute, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Yuandong Feng
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Linlin Zhang
- National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
- Precision Medical Institute, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Ni Guo
- National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Aili He
- National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China.
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China.
| | - Guangyao Kong
- National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China.
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China.
- Precision Medical Institute, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China.
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, Shaanxi, PR China.
| |
Collapse
|
18
|
Wu J, Fan S, Feinberg D, Wang X, Jabbar S, Kang Y. Inhibition of Sphingosine Kinase 2 Results in PARK2-Mediated Mitophagy and Induces Apoptosis in Multiple Myeloma. Curr Oncol 2023; 30:3047-3063. [PMID: 36975444 PMCID: PMC10047154 DOI: 10.3390/curroncol30030231] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/09/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
Mitophagy plays an important role in maintaining mitochondrial homeostasis by clearing damaged mitochondria. Sphingosine kinase 2 (SK2), a type of sphingosine kinase, is an important metabolic enzyme involved in generating sphingosine-1-phosphate. Its expression level is elevated in many cancers and is associated with poor clinical outcomes. However, the relationship between SK2 and mitochondrial dysfunction remains unclear. We found that the genetic downregulation of SK2 or treatment with ABC294640, a specific inhibitor of SK2, induced mitophagy and apoptosis in multiple myeloma cell lines. We showed that mitophagy correlates with apoptosis induction and likely occurs through the SET/PP2AC/PARK2 pathway, where inhibiting PP2AC activity may rescue this process. Furthermore, we found that PP2AC and PARK2 form a complex, suggesting that they might regulate mitophagy through protein-protein interactions. Our study demonstrates the important role of SK2 in regulating mitophagy and provides new insights into the mechanism of mitophagy in multiple myeloma.
Collapse
Affiliation(s)
| | | | | | | | | | - Yubin Kang
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
19
|
Nokkeaw A, Thamjamrassri P, Tangkijvanich P, Ariyachet C. Regulatory Functions and Mechanisms of Circular RNAs in Hepatic Stellate Cell Activation and Liver Fibrosis. Cells 2023; 12:cells12030378. [PMID: 36766720 PMCID: PMC9913196 DOI: 10.3390/cells12030378] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 01/22/2023] Open
Abstract
Chronic liver injury induces the activation of hepatic stellate cells (HSCs) into myofibroblasts, which produce excessive amounts of extracellular matrix (ECM), resulting in tissue fibrosis. If the injury persists, these fibrous scars could be permanent and disrupt liver architecture and function. Currently, effective anti-fibrotic therapies are lacking; hence, understanding molecular mechanisms that control HSC activation could hold a key to the development of new treatments. Recently, emerging studies have revealed roles of circular RNAs (circRNAs), a class of non-coding RNAs that was initially assumed to be the result of splicing errors, as new regulators in HSC activation. These circRNAs can modulate the activity of microRNAs (miRNAs) and their interacting protein partners involved in regulating fibrogenic signaling cascades. In this review, we will summarize the current knowledge of this class of non-coding RNAs for their molecular function in HSC activation and liver fibrosis progression.
Collapse
Affiliation(s)
- Archittapon Nokkeaw
- Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Center of Excellence in Hepatitis and Liver Cancer, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Medical Biochemistry Program, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Pannathon Thamjamrassri
- Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Center of Excellence in Hepatitis and Liver Cancer, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Medical Biochemistry Program, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Pisit Tangkijvanich
- Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Center of Excellence in Hepatitis and Liver Cancer, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Correspondence: (P.T.); (C.A.)
| | - Chaiyaboot Ariyachet
- Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Center of Excellence in Hepatitis and Liver Cancer, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Correspondence: (P.T.); (C.A.)
| |
Collapse
|
20
|
Pang H, Wang J, Wei Q, Liu J, Chu X, Yuan C, Yang B, Li M, Ma D, Tang Y, Wang C, Zhang J. miR-548ag functions as an oncogene by suppressing MOB1B in the development of obesity-related endometrial cancer. Cancer Sci 2022; 114:1507-1518. [PMID: 36445107 PMCID: PMC10067393 DOI: 10.1111/cas.15679] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 11/16/2022] [Accepted: 11/22/2022] [Indexed: 11/30/2022] Open
Abstract
Obesity is a high-risk factor in the development of endometrial cancer (EC). Our previous study observed that miR-548ag was significantly overexpressed in the sera of obese individuals. Here, we report the function of miR-548ag and its mechanism in promoting the obesity-related progression of EC. The content of miR-548ag was increased in the serum of obese EC individuals. Bioinformatics analysis indicated that the survival rate of EC patients with a higher expression of miR-548ag was significantly reduced. The Mps One Binder Kinase Activator 1B (MOB1B, the core member of the Hippo signaling pathway) is a direct target gene of miR-548ag, which is inversely correlated with the expression of miR-548ag. The overexpression of miR-548ag enhances the proliferation, invasion, and migration, and inhibits apoptosis by downregulating the expression of MOB1B, leading to the deactivation of the Hippo pathway in EC cell lines and contributing to tumor progression in vivo. Our study has established that miR-548ag functions as an oncogene by suppressing MOB1B in the development of obesity-related EC.
Collapse
Affiliation(s)
- Huai Pang
- Medical College of Shihezi University, Shihezi, China
| | - Jingzhou Wang
- Medical College of Shihezi University, Shihezi, China
| | - Qianqian Wei
- Medical College of Shihezi University, Shihezi, China
| | - Jie Liu
- Medical College of Shihezi University, Shihezi, China
| | - Xiaolong Chu
- Medical College of Shihezi University, Shihezi, China
| | | | - Bingqi Yang
- Medical College of Shihezi University, Shihezi, China
| | - Menghuan Li
- Medical College of Shihezi University, Shihezi, China
| | - Dingling Ma
- Medical College of Shihezi University, Shihezi, China
| | - Yihan Tang
- Medical College of Shihezi University, Shihezi, China
| | - Cuizhe Wang
- Medical College of Shihezi University, Shihezi, China
| | - Jun Zhang
- Medical College of Shihezi University, Shihezi, China
| |
Collapse
|
21
|
Chen Y, Qian J, Ding P, Wang W, Li X, Tang X, Tang C, Yang Y, Gu C. Elevated SFXN2 limits mitochondrial autophagy and increases iron-mediated energy production to promote multiple myeloma cell proliferation. Cell Death Dis 2022; 13:822. [PMID: 36163342 PMCID: PMC9513108 DOI: 10.1038/s41419-022-05272-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 09/14/2022] [Accepted: 09/15/2022] [Indexed: 01/23/2023]
Abstract
Human sideroflexin 2 (SFXN2) belongs to the SFXN protein family, which is a mitochondrial outer membrane protein involved in mitochondrial iron metabolism. Mitochondria are indispensable for cellular energy production and iron metabolism. However, it remains elusive how SFXN2 modulates mitochondrial homeostasis and cellular iron metabolism in multiple myeloma (MM). In this study, we first found that SFXN2 was significantly elevated and correlated to poor outcomes in MM patients from clinical datasets. SFXN2 overexpression promoted MM cell proliferation and suppressed starvation-induced autophagy/mitophagy, while SFXN2 knockdown aggravated mitochondria damage and autophagic processes in ARP1 and H929 MM cell lines. Furthermore, inhibition of SFXN2 exerted effectively anti-myeloma activity in vivo by using myeloma xenograft model. Mechanism studies indicated that heme oxygenase 1 (HO1) with anti-oxidant function contributed to the process of autophagy suppression and cellular proliferation mediated by SFXN2. Our study revealed the critical role of SFXN2 in regulating mitochondrial bioenergetics, mitophagy, cellular iron metabolism, and redox homeostasis in interconnected and intricate way. Collectively, these findings not only provide insights into the metabolic reprogramming of tumor cells, but also highlight the therapeutic potential of SFXN2 in combination with iron metabolism as target for prognosis and treatment in MM patients.
Collapse
Affiliation(s)
- Ying Chen
- grid.410745.30000 0004 1765 1045Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing, China ,grid.410745.30000 0004 1765 1045School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jinjun Qian
- grid.410745.30000 0004 1765 1045School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Pinggang Ding
- grid.410745.30000 0004 1765 1045School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wang Wang
- grid.410745.30000 0004 1765 1045School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xinying Li
- grid.410745.30000 0004 1765 1045School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaozhu Tang
- grid.410745.30000 0004 1765 1045School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chao Tang
- grid.410745.30000 0004 1765 1045School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ye Yang
- grid.410745.30000 0004 1765 1045School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chunyan Gu
- grid.410745.30000 0004 1765 1045Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing, China ,grid.410745.30000 0004 1765 1045School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
22
|
Xiu X, Zhang H, Xue A, Cooper DN, Yan L, Yang Y, Yang Y, Zhao H. Genetic evidence for a causal relationship between type 2 diabetes and peripheral artery disease in both Europeans and East Asians. BMC Med 2022; 20:300. [PMID: 36042491 PMCID: PMC9429730 DOI: 10.1186/s12916-022-02476-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 07/12/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Observational studies have revealed that type 2 diabetes (T2D) is associated with an increased risk of peripheral artery disease (PAD). However, whether the two diseases share a genetic basis and whether the relationship is causal remain unclear. It is also unclear as to whether these relationships differ between ethnic groups. METHODS By leveraging large-scale genome-wide association study (GWAS) summary statistics of T2D (European-based: Ncase = 21,926, Ncontrol = 342,747; East Asian-based: Ncase = 36,614, Ncontrol = 155,150) and PAD (European-based: Ncase = 5673, Ncontrol = 359,551; East Asian-based: Ncase = 3593, Ncontrol = 208,860), we explored the genetic correlation and putative causal relationship between T2D and PAD in both Europeans and East Asians using linkage disequilibrium score regression and seven Mendelian randomization (MR) models. We also performed multi-trait analysis of GWAS and two gene-based analyses to reveal candidate variants and risk genes involved in the shared genetic basis between T2D and PAD. RESULTS We observed a strong genetic correlation (rg) between T2D and PAD in both Europeans (rg = 0.51; p-value = 9.34 × 10-15) and East Asians (rg = 0.46; p-value = 1.67 × 10-12). The MR analyses provided consistent evidence for a causal effect of T2D on PAD in both ethnicities (odds ratio [OR] = 1.05 to 1.28 for Europeans and 1.15 to 1.27 for East Asians) but not PAD on T2D. This putative causal effect was not influenced by total cholesterol, body mass index, systolic blood pressure, or smoking initiation according to multivariable MR analysis, and the genetic overlap between T2D and PAD was further explored employing an independent European sample through polygenic risk score regression. Multi-trait analysis of GWAS revealed two novel European-specific single nucleotide polymorphisms (rs927742 and rs1734409) associated with the shared genetic basis of T2D and PAD. Gene-based analyses consistently identified one gene ANKFY1 and gene-gene interactions (e.g., STARD10 [European-specific] to AP3S2 [East Asian-specific]; KCNJ11 [European-specific] to KCNQ1 [East Asian-specific]) associated with the trans-ethnic genetic overlap between T2D and PAD, reflecting a common genetic basis for the co-occurrence of T2D and PAD in both Europeans and East Asians. CONCLUSIONS Our study provides the first evidence for a genetically causal effect of T2D on PAD in both Europeans and East Asians. Several candidate variants and risk genes were identified as being associated with this genetic overlap. Our findings emphasize the importance of monitoring PAD status in T2D patients and suggest new genetic biomarkers for screening PAD risk among patients with T2D.
Collapse
Affiliation(s)
- Xuehao Xiu
- Department of Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, China
| | - Haoyang Zhang
- Department of Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, China.,School of Data and Computer Science, Sun Yat-sen University, Guangzhou, 510000, China
| | - Angli Xue
- Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Sydney, NSW, Australia.,Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - David N Cooper
- Institute of Medical Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - Li Yan
- Department of Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, China
| | - Yuedong Yang
- School of Data and Computer Science, Sun Yat-sen University, Guangzhou, 510000, China.
| | - Yuanhao Yang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia. .,Mater Research Institute, Translational Research Institute, Brisbane, QLD, Australia.
| | - Huiying Zhao
- Department of Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, China.
| |
Collapse
|
23
|
Xu ZX, Li JZ, Li Q, Xu MY, Li HY. CircRNA608-microRNA222-PINK1 axis regulates the mitophagy of hepatic stellate cells in NASH related fibrosis. Biochem Biophys Res Commun 2022; 610:35-42. [DOI: 10.1016/j.bbrc.2022.04.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/31/2022] [Accepted: 04/02/2022] [Indexed: 12/11/2022]
|
24
|
Yadav T, Gau D, Roy P. Mitochondria-actin cytoskeleton crosstalk in cell migration. J Cell Physiol 2022; 237:2387-2403. [PMID: 35342955 PMCID: PMC9945482 DOI: 10.1002/jcp.30729] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 03/06/2022] [Accepted: 03/11/2022] [Indexed: 12/15/2022]
Abstract
Mitochondria perform diverse functions in the cell and their roles during processes such as cell survival, differentiation, and migration are increasingly being appreciated. Mitochondrial and actin cytoskeletal networks not only interact with each other, but this multifaceted interaction shapes their functional dynamics. The interrelation between mitochondria and the actin cytoskeleton extends far beyond the requirement of mitochondrial ATP generation to power actin dynamics, and impinges upon several major aspects of cellular physiology. Being situated at the hub of cell signaling pathways, mitochondrial function can alter the activity of actin regulatory proteins and therefore modulate the processes downstream of actin dynamics such as cellular migration. As we will discuss, this regulation is highly nuanced and operates at multiple levels allowing mitochondria to occupy a strategic position in the regulation of migration, as well as pathological events that rely on aberrant cell motility such as cancer metastasis. In this review, we summarize the crosstalk that exists between mitochondria and actin regulatory proteins, and further emphasize on how this interaction holds importance in cell migration in normal as well as dysregulated scenarios as in cancer.
Collapse
Affiliation(s)
- Tarun Yadav
- Department of Biology, Indian Institute of Science Education and Research, Pune, India
| | - David Gau
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Partha Roy
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
25
|
Dai B, Xu J, Li X, Huang L, Hopkins C, Wang H, Yao H, Mi J, Zheng L, Wang J, Tong W, Chow DHK, Li Y, He X, Hu P, Chen Z, Zu H, Li Y, Yao Y, Jiang Q, Qin L. Macrophages in epididymal adipose tissue secrete osteopontin to regulate bone homeostasis. Nat Commun 2022; 13:427. [PMID: 35058428 PMCID: PMC8776868 DOI: 10.1038/s41467-021-27683-w] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 11/04/2021] [Indexed: 12/21/2022] Open
Abstract
Epididymal white adipose tissue (eWAT) secretes an array of cytokines to regulate the metabolism of organs and tissues in high-fat diet (HFD)-induced obesity, but its effects on bone metabolism are not well understood. Here, we report that macrophages in eWAT are a main source of osteopontin, which selectively circulates to the bone marrow and promotes the degradation of the bone matrix by activating osteoclasts, as well as modulating bone marrow-derived macrophages (BMDMs) to engulf the lipid droplets released from adipocytes in the bone marrow of mice. However, the lactate accumulation induced by osteopontin regulation blocks both lipolysis and osteoclastogenesis in BMDMs by limiting the energy regeneration by ATP6V0d2 in lysosomes. Both surgical removal of eWAT and local injection of either clodronate liposomes (for depleting macrophages) or osteopontin-neutralizing antibody show comparable amelioration of HFD-induced bone loss in mice. These results provide an avenue for developing therapeutic strategies to mitigate obesity-related bone disorders.
Collapse
Affiliation(s)
- Bingyang Dai
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Xu Li
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Le Huang
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Chelsea Hopkins
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Honglian Wang
- Research Center for Integrated Medicine, Affiliated Traditional Medicine Hospital of Southwest Medical University, 646000, Luzhou, Sichuan, China
| | - Hao Yao
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Jie Mi
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Lizhen Zheng
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Jiali Wang
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wenxue Tong
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Dick Ho-Kiu Chow
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Ye Li
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Xuan He
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Peijie Hu
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | - Ziyi Chen
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Haiyue Zu
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yixuan Li
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Yao Yao
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Qing Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Ling Qin
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
26
|
Yao J, Wang J, Xu Y, Guo Q, Sun Y, Liu J, Li S, Guo Y, Wei L. CDK9 inhibition blocks the initiation of PINK1-PRKN-mediated mitophagy by regulating the SIRT1-FOXO3-BNIP3 axis and enhances the therapeutic effects involving mitochondrial dysfunction in hepatocellular carcinoma. Autophagy 2021; 18:1879-1897. [PMID: 34890308 PMCID: PMC9450969 DOI: 10.1080/15548627.2021.2007027] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Mitophagy is a type of selective macroautophagy/autophagy that degrades dysfunctional or excessive mitochondria. Regulation of this process is critical for maintaining cellular homeostasis and has been closely implicated in acquired drug resistance. However, the regulatory mechanisms and influences of mitophagy in cancer are still unclear. Here, we reported that inhibition of CDK9 blocked PINK1-PRKN-mediated mitophagy in HCC (hepatocellular carcinoma) by interrupting mitophagy initiation. We demonstrated that CDK9 inhibitors promoted dephosphorylation of SIRT1 and promoted FOXO3 protein degradation, which was regulated by its acetylation, leading to the transcriptional repression of FOXO3-driven BNIP3 and impairing the BNIP3-mediated stability of the PINK1 protein. Lysosomal degradation inhibitors could not rescue mitophagy flux blocked by CDK9 inhibitors. Thus, CDK9 inhibitors inactivated the SIRT1-FOXO3-BNIP3 axis and PINK1-PRKN pathway to subsequently block mitophagy initiation. Moreover, CDK9 inhibitors facilitated mitochondrial dysfunction. The dual effects of CDK9 inhibitors resulted in the destruction of mitochondrial homeostasis and cell death in HCC. Importantly, a novel CDK9 inhibitor, oroxylin A (OA), from Scutellaria baicalensis was investigated, and it showed strong therapeutic potential against HCC and a striking capacity to overcome drug resistance by downregulating PINK1-PRKN-mediated mitophagy. Additionally, because of the moderate and controlled inhibition of CDK9, OA not led to extreme repression of general transcription and appeared to overcome the inconsistent anti-HCC efficacy and high normal tissue toxicity that was associated with existing CDK9 inhibitors. All of the findings reveal that mitophagy disruption is a promising strategy for HCC treatment and OA is a potential candidate for the development of mitophagy inhibitors.Abbreviations: BNIP3: BCL2 interacting protein 3; CCCP: carbonyl cyanide p-trichloromethoxy-phenylhydrazone; CDK9: cyclin dependent kinase 9; CHX: cycloheximide; CQ, chloroquine; DFP: deferiprone; DOX: doxorubicin; EBSS: Earle's balanced salt solution; E64d: aloxistatin; FOXO3: forkhead box O3; HCC: hepatocellular carcinoma; HepG2/ADR: adriamycin-resistant HepG2 cells; MMP: mitochondrial membrane potential; mito-Keima: mitochondria-targeted and pH-sensitive fluorescent protein; MitoSOX: mitochondrial reactive oxygen species; OA: oroxylin A; PB: phosphate buffer; PDX: patient-derived tumor xenograft; PINK1: PTEN induced kinase 1; POLR2A: RNA polymerase II subunit A; p-POLR2A-S2: Ser2 phosphorylation of RNA polymerase II subunit A; PRKN: parkin RBR E3 ubiquitin protein ligase; SIRT1: sirtuin 1.
Collapse
Affiliation(s)
- Jingyue Yao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, The People's Republic of China
| | - Jubo Wang
- Jiangsu Key Laboratory of Drug Design and Optimization, School of Pharmacy, China Pharmaceutical University, Nanjing, The People's Republic of China
| | - Ye Xu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, The People's Republic of China
| | - Qinglong Guo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, The People's Republic of China
| | - Yuening Sun
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, The People's Republic of China
| | - Jian Liu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, The People's Republic of China
| | - Sichan Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, The People's Republic of China
| | - Yongjian Guo
- School of Biopharmacy, China Pharmaceutical University, Nanjing, The People's Republic of China
| | - Libin Wei
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, The People's Republic of China
| |
Collapse
|
27
|
Morciano G, Vezzani B, Missiroli S, Boncompagni C, Pinton P, Giorgi C. An Updated Understanding of the Role of YAP in Driving Oncogenic Responses. Cancers (Basel) 2021; 13:cancers13123100. [PMID: 34205830 PMCID: PMC8234554 DOI: 10.3390/cancers13123100] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/09/2021] [Accepted: 06/17/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary In 2020, the global cancer database GLOBOCAN estimated 19.3 million new cancer cases worldwide. The discovery of targeted therapies may help prognosis and outcome of the patients affected, but the understanding of the plethora of highly interconnected pathways that modulate cell transformation, proliferation, invasion, migration and survival remains an ambitious goal. Here we propose an updated state of the art of YAP as the key protein driving oncogenic response via promoting all those steps at multiple levels. Of interest, the role of YAP in immunosuppression is a field of evolving research and growing interest and this summary about the current pharmacological therapies impacting YAP serves as starting point for future studies. Abstract Yes-associated protein (YAP) has emerged as a key component in cancer signaling and is considered a potent oncogene. As such, nuclear YAP participates in complex and only partially understood molecular cascades that are responsible for the oncogenic response by regulating multiple processes, including cell transformation, tumor growth, migration, and metastasis, and by acting as an important mediator of immune and cancer cell interactions. YAP is finely regulated at multiple levels, and its localization in cells in terms of cytoplasm–nucleus shuttling (and vice versa) sheds light on interesting novel anticancer treatment opportunities and putative unconventional functions of the protein when retained in the cytosol. This review aims to summarize and present the state of the art knowledge about the role of YAP in cancer signaling, first focusing on how YAP differs from WW domain-containing transcription regulator 1 (WWTR1, also named as TAZ) and which upstream factors regulate it; then, this review focuses on the role of YAP in different cancer stages and in the crosstalk between immune and cancer cells as well as growing translational strategies derived from its inhibitory and synergistic effects with existing chemo-, immuno- and radiotherapies.
Collapse
|
28
|
Xu C, Fu Y. Expression Profiles of tRNA-Derived Fragments and Their Potential Roles in Multiple Myeloma. Onco Targets Ther 2021; 14:2805-2814. [PMID: 33911877 PMCID: PMC8075357 DOI: 10.2147/ott.s302594] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 03/16/2021] [Indexed: 01/15/2023] Open
Abstract
Background Multiple myeloma is an incurable hematologic malignancy. The discovery of mechanisms may help to find new therapeutic targets and prolong survival. tRNA-related fragments (tRF) and tRNA halves (tiRNA) are small RNA derived from tRNAs and implicated in a wide variety of pathological processes, including cancer initiation and progression. However, there are almost no research reporting the role of these tRNA derived fragments in myeloma as far as we know. Methods In this study, we proposed the expression profiles of tRFs/tiRNAs in myeloma through RNA-sequencing in new diagnosed myeloma and healthy donors. The expression of selected tRFs/tiRNAs was further validated in clinical specimens by qPCR. Bioinformatic analysis was performed to predict their potential roles in multiple myeloma. Results A total of 33 upregulated tRFs/tiRNAs and 22 downregulated tRFs/tiRNAs were identified. GO enrichment and KEGG pathway analysis were performed to analyze the functions of one significantly up-regulated and one significantly down-regulated tRNA-derived fragments. tRFs/tiRNAs may be involved in MM initiation and progression. Conclusion We concluded that tRFs/tiRNAs were dysregulated and could be potential biomarkers for multiple myeloma.
Collapse
Affiliation(s)
- Cong Xu
- Department of Hematology, The Third Xiangya Hospital of Central South University, Changsha, 410000, People's Republic of China
| | - Yunfeng Fu
- Department of Blood Transfusion, The Third Xiangya Hospital of Central South University, Changsha, 410000, People's Republic of China
| |
Collapse
|
29
|
New Insights into YES-Associated Protein Signaling Pathways in Hematological Malignancies: Diagnostic and Therapeutic Challenges. Cancers (Basel) 2021; 13:cancers13081981. [PMID: 33924049 PMCID: PMC8073623 DOI: 10.3390/cancers13081981] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/03/2021] [Accepted: 04/16/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary YES-associated protein (YAP) is a co-transcriptional activator that binds to transcriptional factors to increase the rate of transcription of a set of genes, and it can intervene in the onset and progression of different tumors. Most of the data in the literature refer to the effects of the YAP system in solid neoplasms. In this review, we analyze the possibility that YAP can also intervene in hematological neoplasms such as lymphomas, multiple myeloma, and acute and chronic leukemias, modifying the phenomena of cell proliferation and cell death. The possibilities of pharmacological intervention related to the YAP system in an attempt to use its modulation therapeutically are also discussed. Abstract The Hippo/YES-associated protein (YAP) signaling pathway is a cell survival and proliferation-control system with its main activity that of regulating cell growth and organ volume. YAP operates as a transcriptional coactivator in regulating the onset, progression, and treatment response in numerous human tumors. Moreover, there is evidence suggesting the involvement of YAP in the control of the hematopoietic system, in physiological conditions rather than in hematological diseases. Nevertheless, several reports have proposed that the effects of YAP in tumor cells are cell-dependent and cell-type-determined, even if YAP usually interrelates with extracellular signaling to stimulate the onset and progression of tumors. In the present review, we report the most recent findings in the literature on the relationship between the YAP system and hematological neoplasms. Moreover, we evaluate the possible therapeutic use of the modulation of the YAP system in the treatment of malignancies. Given the effects of the YAP system in immunosurveillance, tumorigenesis, and chemoresistance, further studies on interactions between the YAP system and hematological malignancies will offer very relevant information for the targeting of these diseases employing YAP modifiers alone or in combination with chemotherapy drugs.
Collapse
|
30
|
Aventaggiato M, Vernucci E, Barreca F, Russo MA, Tafani M. Sirtuins' control of autophagy and mitophagy in cancer. Pharmacol Ther 2020; 221:107748. [PMID: 33245993 DOI: 10.1016/j.pharmthera.2020.107748] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/10/2020] [Indexed: 02/06/2023]
Abstract
Mammalian cells use a specialized and complex machinery for the removal of altered proteins or dysfunctional organelles. Such machinery is part of a mechanism called autophagy. Moreover, when autophagy is specifically employed for the removal of dysfunctional mitochondria, it is called mitophagy. Autophagy and mitophagy have important physiological implications and roles associated with cellular differentiation, resistance to stresses such as starvation, metabolic control and adaptation to the changing microenvironment. Unfortunately, transformed cancer cells often exploit autophagy and mitophagy for sustaining their metabolic reprogramming and growth to a point that autophagy and mitophagy are recognized as promising targets for ongoing and future antitumoral therapies. Sirtuins are NAD+ dependent deacylases with a fundamental role in sensing and modulating cellular response to external stresses such as nutrients availability and therefore involved in aging, oxidative stress control, inflammation, differentiation and cancer. It is clear, therefore, that autophagy, mitophagy and sirtuins share many common aspects to a point that, recently, sirtuins have been linked to the control of autophagy and mitophagy. In the context of cancer, such a control is obtained by modulating transcription of autophagy and mitophagy genes, by post translational modification of proteins belonging to the autophagy and mitophagy machinery, by controlling ROS production or major metabolic pathways such as Krebs cycle or glutamine metabolism. The present review details current knowledge on the role of sirtuins, autophagy and mitophagy in cancer to then proceed to discuss how sirtuins can control autophagy and mitophagy in cancer cells. Finally, we discuss sirtuins role in the context of tumor progression and metastasis indicating glutamine metabolism as an example of how a concerted activation and/or inhibition of sirtuins in cancer cells can control autophagy and mitophagy by impinging on the metabolism of this fundamental amino acid.
Collapse
Affiliation(s)
- Michele Aventaggiato
- Department of Experimental Medicine, Sapienza University, Viale Regina Elena 324, 00161 Rome, Italy
| | - Enza Vernucci
- Department of Internistic, Anesthesiologic and Cardiovascular Clinical Sciences, Italy; MEBIC Consortium, San Raffaele Open University, Via val Cannuta 247, 00166 Rome, Italy
| | - Federica Barreca
- Department of Experimental Medicine, Sapienza University, Viale Regina Elena 324, 00161 Rome, Italy
| | - Matteo A Russo
- MEBIC Consortium, San Raffaele Open University, Via val Cannuta 247, 00166 Rome, Italy; IRCCS San Raffaele, Via val Cannuta 247, 00166 Rome, Italy
| | - Marco Tafani
- Department of Experimental Medicine, Sapienza University, Viale Regina Elena 324, 00161 Rome, Italy.
| |
Collapse
|
31
|
Wang D, He J, Huang B, Liu S, Zhu H, Xu T. Emerging role of the Hippo pathway in autophagy. Cell Death Dis 2020; 11:880. [PMID: 33082313 PMCID: PMC7576599 DOI: 10.1038/s41419-020-03069-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/07/2020] [Accepted: 08/21/2020] [Indexed: 02/06/2023]
Abstract
Autophagy is a dynamic circulatory system that occurs in all eukaryotic cells. Cytoplasmic material is transported to lysosomes for degradation and recovery through autophagy. This provides energy and macromolecular precursors for cell renewal and homeostasis. The Hippo-YAP pathway has significant biological properties in controlling organ size, tissue homeostasis, and regeneration. Recently, the Hippo-YAP axis has been extensively referred to as the pathophysiological processes regulating autophagy. Understanding the cellular and molecular basis of these processes is crucial for identifying disease pathogenesis and novel therapeutic targets. Here we review recent findings from Drosophila models to organisms. We particularly emphasize the regulation between Hippo core components and autophagy, which is involved in normal cellular regulation and the pathogenesis of human diseases, and its application to disease treatment.
Collapse
Affiliation(s)
- Dongying Wang
- Department of Obstetrics and Gynecology, The Second Hospital, Jilin University, 218 Zi Qiang Street, Changchun, Jilin, 130000, China
| | - Jiaxing He
- Department of Obstetrics and Gynecology, The Second Hospital, Jilin University, 218 Zi Qiang Street, Changchun, Jilin, 130000, China
| | - Bingyu Huang
- Department of Obstetrics and Gynecology, The Second Hospital, Jilin University, 218 Zi Qiang Street, Changchun, Jilin, 130000, China
| | - Shanshan Liu
- Department of Obstetrics and Gynecology, The Second Hospital, Jilin University, 218 Zi Qiang Street, Changchun, Jilin, 130000, China
| | - Hongming Zhu
- Department of Obstetrics and Gynecology, The Second Hospital, Jilin University, 218 Zi Qiang Street, Changchun, Jilin, 130000, China
| | - Tianmin Xu
- Department of Obstetrics and Gynecology, The Second Hospital, Jilin University, 218 Zi Qiang Street, Changchun, Jilin, 130000, China.
| |
Collapse
|
32
|
Bortezomib-Loaded Mesoporous Silica Nanoparticles Selectively Alter Metabolism and Induce Death in Multiple Myeloma Cells. Cancers (Basel) 2020; 12:cancers12092709. [PMID: 32967380 PMCID: PMC7565423 DOI: 10.3390/cancers12092709] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/16/2020] [Accepted: 09/17/2020] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Multiple myeloma (MM) is the second most common hematological malignancy and, despite the great advances made in its management, the development of novel therapeutic strategies are still needed in order to extend patients’ survival and to improve their quality of life. Here we show the striking ability of a mesoporous silica-based device to selectively deliver the antineoplastic drug bortezomib to Folate Receptor (FR) overexpressing MM cells, without causing injury nor perturbing the metabolic homeostasis of FR-negative healthy cells. Our data highlight the high efficacy and extraordinary safety of the tested nanodevice, paving the way for its future exploitation in the treatment of MM. Abstract A mesoporous silica-based nanodevice bearing the antineoplastic drug bortezomib (BTZ), whose release is triggered in acidic environment and grafted with folic acid (FOL) as a targeting function (FOL-MSN-BTZ) was tested on folate receptor overexpressing (FR+) multiple myeloma (MM) cells and on FR negative (FR−) normal cells. FOL-MSN-BTZ efficacy studies were conducted by means of growth experiments, TEM, TUNEL assay and Western Blotting analysis (WB). Metabolic investigations were performed to assess cells metabolic response to MSNs treatments. FOL-MSN-BTZ exclusively killed FR+ MM cells, leading to an apoptotic rate that was comparable to that induced by free BTZ, and the effect was accompanied by metabolic dysfunction and oxidative stress. Importantly, FOL-MSN-BTZ treated FR− normal cells did not show any significant sign of injury or metabolic perturbation, while free BTZ was still highly toxic. Notably, the vehicle alone (MSN-FOL) did not affect any biological process in both tested cell models. These data show the striking specificity of FOL-MSN-BTZ toward FR+ tumor cells and the outstanding safety of the MSN-FOL vehicle, paving the way for a future exploitation of FOL-MSN-BTZ in MM target therapy.
Collapse
|