1
|
Rong D, Gao L, Chen Y, Gao XZ, Tang M, Tang H, Gao Y, Lu G, Ling ZQ, Shen HM. Suppression of the LKB1-AMPK-SLC7A11-GSH signaling pathway sensitizes NSCLC to albumin-bound paclitaxel via oxidative stress. Redox Biol 2025; 81:103567. [PMID: 40023979 PMCID: PMC11915006 DOI: 10.1016/j.redox.2025.103567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025] Open
Abstract
Albumin-bound paclitaxel (nab-PTX) is an important chemotherapeutic drug used for the treatment of advanced and metastatic non-small cell lung cancer (NSCLC). One critical issue in its clinical application is the development of resistance; thus, a deeper understanding of the mechanisms underlying the primary resistance to nab-PTX is expected to help to develop effective therapeutic strategies to overcome resistance. In this study, we made an unexpected discovery that NSCLC with wild-type (WT) Liver kinase B1 (LKB1), an important tumor suppressor and upstream kinase of AMP-activated protein kinase (AMPK), is more resistant to nab-PTX than NSCLC with mutant LKB1. Mechanistically, LKB1 status does not alter the intracellular concentration of nab-PTX or affect its canonical pharmacological action in promoting microtubule polymerization. Instead, we found that LKB1 mediates AMPK activation, leading to increased expression of SLC7A11, a key amino acid transporter and intracellular level of glutathione (GSH), which then attenuates the production of reactive oxygen species (ROS) and apoptotic cell death induced by nab-PTX. On the other hand, genetic or pharmacological inhibition of AMPK in LKB1-WT NSCLC reduces the expression of SLC7A11 and intracellular GSH, increases ROS level, and eventually promotes the apoptotic cell death induced by nab-PTX in vitro. Consistently, the combination of nab-PTX with an AMPK inhibitor exhibits a greater therapeutic efficacy in LKB1-WT NSCLC using xenograft models in vivo. Taken together, our data reveal a novel role of LKB1-AMPK-SLC7A11-GSH signaling pathway in the primary resistance to nab-PTX, and provide a therapeutic strategy for the treatment of LKB1-WT NSCLC by targeting the LKB1-AMPK-SLC7A11-GSH pathway.
Collapse
Affiliation(s)
- Dade Rong
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Liangliang Gao
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Yiguan Chen
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Xiang-Zheng Gao
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Mingzhu Tang
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Haimei Tang
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China; Department of Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Yuan Gao
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Guang Lu
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhi-Qiang Ling
- Experimental Research Centre, The Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Han-Ming Shen
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China.
| |
Collapse
|
2
|
Matsumoto K, Matsumoto Y, Wada J. PARylation-mediated post-transcriptional modifications in cancer immunity and immunotherapy. Front Immunol 2025; 16:1537615. [PMID: 40134437 PMCID: PMC11933034 DOI: 10.3389/fimmu.2025.1537615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 02/17/2025] [Indexed: 03/27/2025] Open
Abstract
Poly-ADP-ribosylation (PARylation) is a post-translational modification in which ADP-ribose is added to substrate proteins. PARylation is mediated by a superfamily of ADP-ribosyl transferases known as PARPs and influences a wide range of cellular functions, including genome integrity maintenance, and the regulation of proliferation and differentiation. We and others have recently reported that PARylation of SH3 domain-binding protein 2 (3BP2) plays a role in bone metabolism, immune system regulation, and cytokine production. Additionally, PARylation has recently gained attention as a target for cancer treatment. In this review, we provide an overview of PARylation, its involvement in several signaling pathways related to cancer immunity, and the potential of combination therapies with PARP inhibitors and immune checkpoint inhibitors.
Collapse
Affiliation(s)
| | - Yoshinori Matsumoto
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Faculty of
Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | | |
Collapse
|
3
|
Tomas EJ, Valdes YR, Davis J, Kolendowski B, Buensuceso A, DiMattia GE, Shepherd TG. Exploiting Cancer Dormancy Signaling Mechanisms in Epithelial Ovarian Cancer Through Spheroid and Organoid Analysis. Cells 2025; 14:133. [PMID: 39851561 PMCID: PMC11764263 DOI: 10.3390/cells14020133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 01/26/2025] Open
Abstract
Epithelial ovarian cancer (EOC) exhibits a unique mode of metastasis, involving spheroid formation in the peritoneum. Our research on EOC spheroid cell biology has provided valuable insights into the signaling plasticity associated with metastasis. We speculate that EOC cells modify their biology between tumour and spheroid states during cancer dormancy, although the specific mechanisms underlying this transition remain unknown. Here, we present novel findings from direct comparisons between cultured EOC spheroids and organoids. Our results indicated that AMP-activated protein kinase (AMPK) activity was significantly upregulated and protein kinase B (Akt) was downregulated in EOC spheroids compared to organoids, suggesting a clear differential phenotype. Through RNA sequencing analysis, we further supported these phenotypic differences and highlighted the significance of cell cycle regulation in organoids. By inhibiting the G2/M checkpoint via kinase inhibitors, we confirmed that this pathway is essential for organoids. Interestingly, our results suggest that specifically targeting aurora kinase A (AURKA) may represent a promising therapeutic strategy since our cells were equally sensitive to Alisertib treatment as both spheroids and organoids. Our findings emphasize the importance of studying cellular adaptations of EOC cells, as there may be different therapeutic targets depending on the step of EOC disease progression.
Collapse
Affiliation(s)
- Emily J. Tomas
- The Mary and John Knight Translational Ovarian Cancer Research Unit, Verspeeten Family Cancer Centre, London, ON N6A 5W9, Canada
- Department of Anatomy & Cell Biology, Western University, London, ON N6A 5C1, Canada
| | - Yudith Ramos Valdes
- The Mary and John Knight Translational Ovarian Cancer Research Unit, Verspeeten Family Cancer Centre, London, ON N6A 5W9, Canada
| | - Jennifer Davis
- The Mary and John Knight Translational Ovarian Cancer Research Unit, Verspeeten Family Cancer Centre, London, ON N6A 5W9, Canada
| | - Bart Kolendowski
- The Mary and John Knight Translational Ovarian Cancer Research Unit, Verspeeten Family Cancer Centre, London, ON N6A 5W9, Canada
| | - Adrian Buensuceso
- The Mary and John Knight Translational Ovarian Cancer Research Unit, Verspeeten Family Cancer Centre, London, ON N6A 5W9, Canada
| | - Gabriel E. DiMattia
- The Mary and John Knight Translational Ovarian Cancer Research Unit, Verspeeten Family Cancer Centre, London, ON N6A 5W9, Canada
- Department of Oncology, Western University, London, ON N6A 5C1, Canada
- Department of Biochemistry, Western University, London, ON N6A 5C1, Canada
| | - Trevor G. Shepherd
- The Mary and John Knight Translational Ovarian Cancer Research Unit, Verspeeten Family Cancer Centre, London, ON N6A 5W9, Canada
- Department of Anatomy & Cell Biology, Western University, London, ON N6A 5C1, Canada
- Department of Oncology, Western University, London, ON N6A 5C1, Canada
- Department of Obstetrics & Gynaecology, Western University, London, ON N6A 5C1, Canada
| |
Collapse
|
4
|
Kang J, Gallucci S, Pan J, Oakhill JS, Sanij E. The role of STK11/LKB1 in cancer biology: implications for ovarian tumorigenesis and progression. Front Cell Dev Biol 2024; 12:1449543. [PMID: 39544365 PMCID: PMC11560430 DOI: 10.3389/fcell.2024.1449543] [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: 06/15/2024] [Accepted: 10/11/2024] [Indexed: 11/17/2024] Open
Abstract
STK11 (serine-threonine kinase 11), also known as LKB1 (liver kinase B1) is a highly conserved master kinase that regulates cellular metabolism and polarity through a complex signaling network involving AMPK and 12 other AMPK-related kinases. Germline mutations in LKB1 have been causatively linked to Peutz-Jeghers Syndrome (PJS), an autosomal dominant hereditary disease with high cancer susceptibility. The identification of inactivating somatic mutations in LKB1 in different types of cancer further supports its tumor suppressive role. Deleterious mutations in LKB1 are frequently observed in patients with epithelial ovarian cancer. However, its inconsistent effects on tumorigenesis and cancer progression suggest that its functional impact is genetic context-dependent, requiring cooperation with other oncogenic lesions. In this review, we summarize the pleiotropic functions of LKB1 and how its altered activity in cancer cells is linked to oncogenic proliferation and growth, metastasis, metabolic reprogramming, genomic instability, and immune modulation. We also review the current mechanistic understandings of this master kinase as well as therapeutic implications with particular focus on the effects of LKB1 deficiency in ovarian cancer pathogenesis. Lastly, we discuss whether LKB1 deficiency can be exploited as an Achilles heel in ovarian cancer.
Collapse
Affiliation(s)
- Jian Kang
- St Vincent’s Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medicine-St Vincent’s Hospital, University of Melbourne, Melbourne, VIC, Australia
| | - Stefano Gallucci
- St Vincent’s Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medicine-St Vincent’s Hospital, University of Melbourne, Melbourne, VIC, Australia
| | - Junqi Pan
- St Vincent’s Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medicine-St Vincent’s Hospital, University of Melbourne, Melbourne, VIC, Australia
| | - Jonathan S. Oakhill
- St Vincent’s Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medicine-St Vincent’s Hospital, University of Melbourne, Melbourne, VIC, Australia
| | - Elaine Sanij
- St Vincent’s Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medicine-St Vincent’s Hospital, University of Melbourne, Melbourne, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| |
Collapse
|
5
|
Hou J, Mei K, Wang D, Ke S, Chen X, Shang J, Li G, Gao Y, Xiong H, Zhang H, Chen L, Zhang W, Deng Y, Hong X, Liu DA, Hu T, Guo W, Zhan YY. TGM1/3-mediated transamidation of Exo70 promotes tumor metastasis upon LKB1 inactivation. Cell Rep 2024; 43:114604. [PMID: 39146185 DOI: 10.1016/j.celrep.2024.114604] [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: 10/04/2023] [Revised: 05/10/2024] [Accepted: 07/24/2024] [Indexed: 08/17/2024] Open
Abstract
Exo70, a key exocyst complex component, is crucial for cell motility and extracellular matrix (ECM) remodeling in cancer metastasis. Despite its potential as a drug target, Exo70's post-translational modifications (PTMs) are poorly characterized. Here, we report that Exo70 is transamidated on Gln5 with Lys56 of cystatin A by transglutaminases TGM1 and TGM3, promoting tumor metastasis. This modification enhances Exo70's association with other exocyst subunits, essential for secreting matrix metalloproteinases, forming invadopodia, and delivering integrins to the leading edge. Tumor suppressor liver kinase B1 (LKB1), whose inactivation accelerates metastasis, phosphorylates TGM1 and TGM3 at Thr386 and Thr282, respectively, to inhibit their interaction with Exo70 and the following transamidation. Cantharidin, a US Food and Drug Administration (FDA)-approved drug, inhibits Exo70 transamidation to restrain tumor cell migration and invasion. Together, our findings highlight Exo70 transamidation as a key molecular mechanism and target and propose cantharidin as a therapeutic strategy with direct clinical translational value for metastatic cancers, especially those with LKB1 loss.
Collapse
Affiliation(s)
- Jihuan Hou
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Kunrong Mei
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Daxuan Wang
- Department of Respiratory Medicine, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Sunkui Ke
- Department of Thoracic Surgery, Zhongshan Hospital Affiliated to Xiamen University, Xiamen 361004, China
| | - Xiong Chen
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Jin Shang
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Guixia Li
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Yan Gao
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Huifang Xiong
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Haoran Zhang
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Lu Chen
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Wenqing Zhang
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Yabin Deng
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Xiaoting Hong
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Di-Ao Liu
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tianhui Hu
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China.
| | - Wei Guo
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Yan-Yan Zhan
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
6
|
Chen G, Li MY, Yang JY, Zhou ZH. Will AMPK be a potential therapeutic target for hepatocellular carcinoma? Am J Cancer Res 2024; 14:3241-3258. [PMID: 39113872 PMCID: PMC11301289 DOI: 10.62347/yavk1315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 07/02/2024] [Indexed: 08/10/2024] Open
Abstract
Cancer is the disease that poses the greatest threat to human health today. Among them, hepatocellular carcinoma (HCC) is particularly prominent due to its high recurrence rate and extremely low five-year postoperative survival rate. In addition to surgical treatment, radiotherapy, chemotherapy, and immunotherapy are the main methods for treating HCC. Due to the natural drug resistance of chemoradiotherapy and targeted drugs, satisfactory results have not been achieved in terms of therapeutic efficacy and cost. AMP-Activated Protein Kinase (AMPK) is a serine/threonine protein kinase. It mainly coordinates the metabolism and transformation of energy between cells, which maintaining a balance between energy supply and demand. The processes of cell growth, proliferation, autophagy, and survival all involve various reaction of cells to energy changes. The regulatory role of AMPK in cellular energy metabolism plays an important role in the occurrence, development, treatment, and prognosis of HCC. Here, we reviewed the latest progress on the regulatory role of AMPK in the occurrence and development of HCC. Firstly, the molecular structure and activation mechanism of AMPK were introduced. Secondly, the emerging regulator related to AMPK and tumors were elaborated. Next, the multitasking roles of AMPK in the occurrence and development mechanism of HCC were discussed separately. Finally, the translational implications and the challenges of AMPK-targeted therapies for HCC treatment were elaborated. In summary, these pieces of information suggest that AMPK can serve as a promising specific therapeutic target for the treatment of HCC.
Collapse
Affiliation(s)
- Guo Chen
- Department of Oncology, Anhui Hospital, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese MedicineHefei, Anhui, China
| | - Ming-Yuan Li
- Department of Oncology, Anhui Hospital, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese MedicineHefei, Anhui, China
| | - Jing-Yi Yang
- Department of Oncology, Feixi Hospital of Traditional Chinese MedicineFeixi, Hefei, Anhui, China
| | - Zhen-Hua Zhou
- Department of Hepatopathy, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese MedicineShanghai, China
| |
Collapse
|
7
|
Lee YS, Im J, Yang Y, Lee HJ, Lee MR, Woo SM, Park SJ, Kong SY, Kim JY, Hwang H, Kim YH. New Function Annotation of PROSER2 in Pancreatic Ductal Adenocarcinoma. J Proteome Res 2024; 23:905-915. [PMID: 38293943 PMCID: PMC10913870 DOI: 10.1021/acs.jproteome.3c00632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/09/2024] [Accepted: 01/15/2024] [Indexed: 02/01/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a dismal prognosis due to the absence of diagnostic markers and molecular targets. Here, we took an unconventional approach to identify new molecular targets for pancreatic cancer. We chose uncharacterized protein evidence level 1 without function annotation from extensive proteomic research on pancreatic cancer and focused on proline and serine-rich 2 (PROSER2), which ranked high in the cell membrane and cytoplasm. In our study using cell lines and patient-derived orthotopic xenograft cells, PROSER2 exhibited a higher expression in cells derived from primary tumors than in those from metastatic tissues. PROSER2 was localized in the cell membrane and cytosol by immunocytochemistry. PROSER2 overexpression significantly reduced the metastatic ability of cancer cells, whereas its suppression had the opposite effect. Proteomic analysis revealed that PROSER2 interacts with STK25 and PDCD10, and their binding was confirmed by immunoprecipitation and immunocytochemistry. STK25 knockdown enhanced metastasis by decreasing p-AMPK levels, whereas PROSER2-overexpressing cells increased the level of p-AMPK, indicating that PROSER2 suppresses invasion via the AMPK pathway by interacting with STK25. This is the first demonstration of the novel role of PROSER2 in antagonizing tumor progression via the STK25-AMPK pathway in PDAC. LC-MS/MS data are available at MassIVE (MSV000092953) and ProteomeXchange (PXD045646).
Collapse
Affiliation(s)
- Yu-Sun Lee
- Division
of Convergence Technology, Research Institute
of National Cancer Center, Goyang 10408, Republic
of Korea
- Department
of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic
of Korea
| | - Jieun Im
- Division
of Convergence Technology, Research Institute
of National Cancer Center, Goyang 10408, Republic
of Korea
| | - Yeji Yang
- Research
Center for Bioconvergence Analysis, Korea
Basic Science Institute, Cheongju 28119, Republic
of Korea
- Critical
Diseases Diagnostics Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Hea Ji Lee
- Research
Center for Bioconvergence Analysis, Korea
Basic Science Institute, Cheongju 28119, Republic
of Korea
| | - Mi Rim Lee
- Department
of Cancer Biomedical Science, National Cancer
Center Graduate School of Cancer Science and Policy, Goyang 10408, Republic of Korea
| | - Sang-Myung Woo
- Department
of Cancer Biomedical Science, National Cancer
Center Graduate School of Cancer Science and Policy, Goyang 10408, Republic of Korea
- Department
of Center for Liver and Pancreatobiliary Cancer, National Cancer Center, Goyang 10408, Republic
of Korea
| | - Sang-Jae Park
- Department
of Center for Liver and Pancreatobiliary Cancer, National Cancer Center, Goyang 10408, Republic
of Korea
- Department
of Surgical Oncology Branch, Research Institute
of National Cancer Center, Goyang 10408, Republic
of Korea
| | - Sun-Young Kong
- Department
of Cancer Biomedical Science, National Cancer
Center Graduate School of Cancer Science and Policy, Goyang 10408, Republic of Korea
- Department
of Targeted Therapy Branch, Research Institute
of National Cancer Center, Goyang 10408, Republic
of Korea
| | - Jin Young Kim
- Research
Center for Bioconvergence Analysis, Korea
Basic Science Institute, Cheongju 28119, Republic
of Korea
- Critical
Diseases Diagnostics Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Heeyoun Hwang
- Research
Center for Bioconvergence Analysis, Korea
Basic Science Institute, Cheongju 28119, Republic
of Korea
- Critical
Diseases Diagnostics Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Yun-Hee Kim
- Division
of Convergence Technology, Research Institute
of National Cancer Center, Goyang 10408, Republic
of Korea
- Department
of Cancer Biomedical Science, National Cancer
Center Graduate School of Cancer Science and Policy, Goyang 10408, Republic of Korea
| |
Collapse
|
8
|
Bosso M, Haddad D, Al Madhoun A, Al-Mulla F. Targeting the Metabolic Paradigms in Cancer and Diabetes. Biomedicines 2024; 12:211. [PMID: 38255314 PMCID: PMC10813379 DOI: 10.3390/biomedicines12010211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/09/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Dysregulated metabolic dynamics are evident in both cancer and diabetes, with metabolic alterations representing a facet of the myriad changes observed in these conditions. This review delves into the commonalities in metabolism between cancer and type 2 diabetes (T2D), focusing specifically on the contrasting roles of oxidative phosphorylation (OXPHOS) and glycolysis as primary energy-generating pathways within cells. Building on earlier research, we explore how a shift towards one pathway over the other serves as a foundational aspect in the development of cancer and T2D. Unlike previous reviews, we posit that this shift may occur in seemingly opposing yet complementary directions, akin to the Yin and Yang concept. These metabolic fluctuations reveal an intricate network of underlying defective signaling pathways, orchestrating the pathogenesis and progression of each disease. The Warburg phenomenon, characterized by the prevalence of aerobic glycolysis over minimal to no OXPHOS, emerges as the predominant metabolic phenotype in cancer. Conversely, in T2D, the prevailing metabolic paradigm has traditionally been perceived in terms of discrete irregularities rather than an OXPHOS-to-glycolysis shift. Throughout T2D pathogenesis, OXPHOS remains consistently heightened due to chronic hyperglycemia or hyperinsulinemia. In advanced insulin resistance and T2D, the metabolic landscape becomes more complex, featuring differential tissue-specific alterations that affect OXPHOS. Recent findings suggest that addressing the metabolic imbalance in both cancer and diabetes could offer an effective treatment strategy. Numerous pharmaceutical and nutritional modalities exhibiting therapeutic effects in both conditions ultimately modulate the OXPHOS-glycolysis axis. Noteworthy nutritional adjuncts, such as alpha-lipoic acid, flavonoids, and glutamine, demonstrate the ability to reprogram metabolism, exerting anti-tumor and anti-diabetic effects. Similarly, pharmacological agents like metformin exhibit therapeutic efficacy in both T2D and cancer. This review discusses the molecular mechanisms underlying these metabolic shifts and explores promising therapeutic strategies aimed at reversing the metabolic imbalance in both disease scenarios.
Collapse
Affiliation(s)
- Mira Bosso
- Department of Pathology, Faculty of Medicine, Health Science Center, Kuwait University, Safat 13110, Kuwait
| | - Dania Haddad
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (D.H.); (A.A.M.)
| | - Ashraf Al Madhoun
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (D.H.); (A.A.M.)
- Department of Animal and Imaging Core Facilities, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Fahd Al-Mulla
- Department of Pathology, Faculty of Medicine, Health Science Center, Kuwait University, Safat 13110, Kuwait
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (D.H.); (A.A.M.)
| |
Collapse
|
9
|
Li GZ, Meng GX, Pan GQ, Zhang X, Yan LJ, Li RZ, Ding ZN, Tan SY, Wang DX, Tian BW, Yan YC, Dong ZR, Hong JG, Li T. MALAT1/ mir-1-3p mediated BRF2 expression promotes HCC progression via inhibiting the LKB1/AMPK signaling pathway. Cancer Cell Int 2023; 23:188. [PMID: 37653482 PMCID: PMC10472681 DOI: 10.1186/s12935-023-03034-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/19/2023] [Indexed: 09/02/2023] Open
Abstract
BACKGROUND The long non-coding RNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) has been reported to play a vital role in the occurrence and development of various tumors. However, the underlying mechanism of MALAT1 in hepatocellular carcinoma (HCC) has not been thoroughly elucidated. METHODS The expression levels of MALAT1 in HCC tissues and different cell lines were detected by qRT-PCR. Antisense oligonucleotides (ASO)-MALAT1 transfected cells were used to explore the biological effects of MALAT1 in HCC cells by cell counting kit 8 (CCK-8), colony formation, transwell, wound healing, and flow cytometry analysis. Western blotting was performed to measure AMPK and apoptosis-related protein levels. Dual-luciferase reporter assay was performed to verify the relationship between MALAT1 and its specific targets. RESULTS We found that MALAT1 was upregulated in HCC, and MALAT1 knockdown in HCC cells inhibited cell proliferation, migration, and invasion and inhibited apoptosis in vitro. Further studies demonstrated that MALAT1 positively regulated the expression of transcription factor II B‑related factor 2 (BRF2), which was associated with tumor recurrence, large tumor size, and poor prognosis in HCC. Mechanistically, MALAT1 was found to act as a competitive endogenous RNA to sponge has-miR-1-3p, which upregulated BRF2 expression. Knockdown of BRF2 inhibited the progression of HCC by activating the LKB1/AMPK signaling pathway. Overexpression of BRF2 reversed the inhibitory effect of MALAT1 knockdown on HCC cell viability. Moreover, ASO targeting MALAT1 inhibited the growth of xenograft tumors. CONCLUSIONS Our results demonstrate a novel MALAT1/miR-1-3p/BRF2/LKB1/AMPK regulatory axis in HCC, which may provide new molecular therapeutic targets for HCC in the future.
Collapse
Affiliation(s)
- Guang-Zhen Li
- Medical Integration and Practice Center, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China
| | - Guang-Xiao Meng
- Department of General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China
- Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Guo-Qiang Pan
- Department of General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China
- Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Xiao Zhang
- Department of General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China
- Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Lun-Jie Yan
- Department of General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China
- Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Rui-Zhe Li
- Department of General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China
- Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Zi-Niu Ding
- Department of General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China
| | - Si-Yu Tan
- Department of General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China
| | - Dong-Xu Wang
- Department of General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China
| | - Bao-Wen Tian
- Department of General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China
| | - Yu-Chuan Yan
- Department of General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China
- Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Zhao-Ru Dong
- Department of General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China
| | - Jian-Guo Hong
- Department of General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China.
| | - Tao Li
- Department of General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China.
| |
Collapse
|
10
|
Lee YCG, Chou FN, Tung SY, Chou HC, Ko TL, Fann YC, Juan SH. Tumoricidal Activity of Simvastatin in Synergy with RhoA Inactivation in Antimigration of Clear Cell Renal Cell Carcinoma Cells. Int J Mol Sci 2023; 24:ijms24119738. [PMID: 37298689 DOI: 10.3390/ijms24119738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 05/30/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023] Open
Abstract
Among kidney cancers, clear cell renal cell carcinoma (ccRCC) has the highest incidence rate in adults. The survival rate of patients diagnosed as having metastatic ccRCC drastically declines even with intensive treatment. We examined the efficacy of simvastatin, a lipid-lowering drug with reduced mevalonate synthesis, in ccRCC treatment. Simvastatin was found to reduce cell viability and increase autophagy induction and apoptosis. In addition, it reduced cell metastasis and lipid accumulation, the target proteins of which can be reversed through mevalonate supplementation. Moreover, simvastatin suppressed cholesterol synthesis and protein prenylation that is essential for RhoA activation. Simvastatin might also reduce cancer metastasis by suppressing the RhoA pathway. A gene set enrichment analysis (GSEA) of the human ccRCC GSE53757 data set revealed that the RhoA and lipogenesis pathways are activated. In simvastatin-treated ccRCC cells, although RhoA was upregulated, it was mainly restrained in the cytosolic fraction and concomitantly reduced Rho-associated protein kinase activity. RhoA upregulation might be a negative feedback effect owing to the loss of RhoA activity caused by simvastatin, which can be restored by mevalonate. RhoA inactivation by simvastatin was correlated with decreased cell metastasis in the transwell assay, which was mimicked in dominantly negative RhoA-overexpressing cells. Thus, owing to the increased RhoA activation and cell metastasis in the human ccRCC dataset analysis, simvastatin-mediated Rho inactivation might serve as a therapeutic target for ccRCC patients. Altogether, simvastatin suppressed the cell viability and metastasis of ccRCC cells; thus, it is a potentially effective ccRCC adjunct therapy after clinical validation for ccRCC treatment.
Collapse
Affiliation(s)
- Yuan-Chii Gladys Lee
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Fang-Ning Chou
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Szu-Yu Tung
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Hsiu-Chu Chou
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Tsui-Ling Ko
- College of Science, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan
| | - Yang C Fann
- Intramural IT and Bioinformatics Program, Division of Intramural, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Shu-Hui Juan
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
11
|
Tomas E, Shepherd TG. Insights into high-grade serous carcinoma pathobiology using three-dimensional culture model systems. J Ovarian Res 2023; 16:70. [PMID: 37038202 PMCID: PMC10088149 DOI: 10.1186/s13048-023-01145-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 03/28/2023] [Indexed: 04/12/2023] Open
Abstract
Epithelial ovarian cancer (EOC) research has become more complex as researchers try to fully understand the metastatic process. Especially as we delve into the concept of tumour dormancy, where cells transition between proliferative and dormant states to survive during disease progression. Thus, the in vitro models used to conduct this research need to reflect this vast biological complexity. The innovation behind the many three-dimensional (3D) spheroid models has been refined to easily generate reproducible spheroids so that we may understand the various molecular signaling changes of cells during metastasis and determine therapeutic efficacy of treatments. This ingenuity was then used to develop the 3D ex vivo patient-derived organoid model, as well as multiple co-culture model systems for EOC research. Although, researchers need to continue to push the boundaries of these current models for in vitro and even in vivo work in the future. In this review, we describe the 3D models already in use, where these models can be developed further and how we can use these models to gain the most knowledge on EOC pathogenesis and discover new targeted therapies.
Collapse
Affiliation(s)
- Emily Tomas
- London Regional Cancer Program, The Mary & John Knight Translational Ovarian Cancer Research Unit, 790 Commissioners Rd. E. Room A4-836, London, ON, N6A 4L6, Canada
- Department of Anatomy & Cell Biology, Western University, London, ON, Canada
| | - Trevor G Shepherd
- London Regional Cancer Program, The Mary & John Knight Translational Ovarian Cancer Research Unit, 790 Commissioners Rd. E. Room A4-836, London, ON, N6A 4L6, Canada.
- Department of Anatomy & Cell Biology, Western University, London, ON, Canada.
- Department of Obstetrics & Gynaecology, Western University, London, ON, Canada.
- Department of Oncology, Western University, London, ON, Canada.
| |
Collapse
|
12
|
Metabolic Regulation of T cell Activity: Implications for Metabolic-Based T-cell Therapies for Cancer. IRANIAN BIOMEDICAL JOURNAL 2023; 27:1-14. [PMID: 36624636 PMCID: PMC9971708 DOI: 10.52547/ibj.3811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Immunometabolism is an emerging field in tumor immunotherapy. Understanding the metabolic competition for access to the limited nutrients between tumor cells and immune cells can reveal the complexity of the tumor microenvironment and help develop new therapeutic approaches for cancer. Recent studies have focused on modifying the function of immune cells by manipulating their metabolic pathways. Besides, identifying metabolic events, which affect the function of immune cells leads to new therapeutic opportunities for treatment of inflammatory diseases and immune-related conditions. According to the literature, metabolic pathway such as glycolysis, tricarboxylic acid cycle, and fatty acid metabolism, significantly influence the survival, proliferation, activation, and function of immune cells and thus regulate immune responses. In this paper, we reviewed the role of metabolic processes and major signaling pathways involving in T-cell regulation and T-cell responses against tumor cells. Moreover, we summarized the new therapeutics suggested to enhance anti-tumor activity of T cells through manipulating metabolic pathways.
Collapse
|
13
|
Chen Y, Peng C, Tan W, Yu J, Zayas J, Peng Y, Lou Z, Pei H, Wang L. Tumor protein D52 (TPD52) affects cancer cell metabolism by negatively regulating AMPK. Cancer Med 2023; 12:488-499. [PMID: 35666017 PMCID: PMC9844640 DOI: 10.1002/cam4.4911] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 04/18/2022] [Accepted: 05/25/2022] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND The AMP-activated protein kinase (AMPK) is a central regulator of energy homeostasis, with deregulation leading to cancer and other diseases. However, how this pathway is dysregulated in cancer has not been well clarified. METHODS Using a tandem affinity purification/mass-spec technique and biochemical analyses, we identified tumor protein D52 (TPD52) as an AMPKα-interacting molecule. To explore the biological effects of TPD52 in cancers, we conducted biochemical and metabolic assays in vitro and in vivo with cancer cells and TPD52 transgenic mice. Finally, we assessed the clinical significance of TPD52 expression in breast cancer patients using bioinformatics techniques. RESULTS TPD52, initially identified to be overexpressed in many human cancers, was found to form a stable complex with AMPK in cancer cells. TPD52 directly interacts with AMPKα and inhibits AMPKα kinase activity in vitro and in vivo. In TPD52 transgenic mice, overexpression of TPD52 leads to AMPK inhibition and multiple metabolic defects. Clinically, high TPD52 expression predicts poor survival of breast cancer patients. CONCLUSION The findings revealed that TPD52 is a novel regulator of energy stress-induced AMPK activation and cell metabolism. These results shed new light on AMPK regulation and understanding of the etiology of cancers with TPD52 overexpression.
Collapse
Affiliation(s)
- Yali Chen
- Department of OncologyGeorgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical CenterWashingtonDistrict of ColumbiaUSA
| | - Changmin Peng
- Department of Biochemistry and Molecular MedicineThe George Washington University School of Medicine and Health ScienceWashingtonDistrict of ColumbiaUSA
| | - Wei Tan
- Department of OncologyGeorgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical CenterWashingtonDistrict of ColumbiaUSA
- Department of Biochemistry and Molecular MedicineThe George Washington University School of Medicine and Health ScienceWashingtonDistrict of ColumbiaUSA
| | - Jia Yu
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo ClinicRochesterMinnesotaUSA
| | - Jacqueline Zayas
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo ClinicRochesterMinnesotaUSA
| | - Yihan Peng
- Department of OncologyGeorgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical CenterWashingtonDistrict of ColumbiaUSA
| | - Zhenkun Lou
- Division of Oncology Research, Department of Oncology, Mayo ClinicRochesterMinnesotaUSA
| | - Huadong Pei
- Department of OncologyGeorgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical CenterWashingtonDistrict of ColumbiaUSA
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo ClinicRochesterMinnesotaUSA
| |
Collapse
|
14
|
Yun H, Han GH, Kim J, Chung J, Kim J, Cho H. NANOG
regulates epithelial–mesenchymal transition via
AMPK
/
mTOR
signalling pathway in ovarian cancer
SKOV
‐3 and
A2780
cells. J Cell Mol Med 2022; 26:5277-5291. [PMID: 36114703 PMCID: PMC9575063 DOI: 10.1111/jcmm.17557] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/26/2022] [Accepted: 09/02/2022] [Indexed: 11/28/2022] Open
Abstract
NANOG engages with tumour initiation and metastasis by regulating the epithelial–mesenchymal transition (EMT) in epithelial ovarian cancer (EOC). However, its role in association with pAMPKα, and its clinical significance in EOC have not been elucidated even though AMPK is known to degrade NANOG in various human cancers. Hence, we investigated the role of pAMPKα and its association with NANOG as potential prognostic biomarkers in EOC. Both NANOG and pAMPKα expression were significantly overexpressed in EOCs comparing nonadjacent normal epithelial tissues, benign tissues, and borderline tumours. NANOG overexpression was significantly associated with poor disease‐free survival (DFS) and overall survival (OS), whereas pAMPKα overexpression was associated with good DFS and OS. Importantly, multivariate analysis revealed that the combination of high NANOG and low pAMPKα expression was a poor independent prognostic factor for DFS and was associated with platinum resistance. In ovarian cancer cell lines, siRNA‐mediated NANOG knockdown diminished migration and invasion properties by regulating the EMT process via the AMPK/mTOR signalling pathway. Furthermore, treatment with AMPK activator suppressed expression of stemness factors such as NANOG, Oct4 and Sox2. Collectively, these findings established that the combination of high NANOG and low pAMPKα expression was associated with EOC progression and platinum resistance, suggesting a potential prognostic biomarker for clinical management in EOC patients.
Collapse
Affiliation(s)
- Hee Yun
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital Yonsei University College of Medicine Seoul Korea
| | - Gwan Hee Han
- Department of Obstetrics and Gynecology Kyung Hee University Hospital at Gangdong Seoul Korea
| | - Julie Kim
- Weill Cornell Medical College New York New York USA
| | - Joon‐Yong Chung
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute National Institutes of Health Bethesda Maryland USA
| | - Jae‐Hoon Kim
- Department of Obstetrics and Gynecology Yonsei University College of Medicine Seoul Korea
- Institute of Women's Life Medical Science Yonsei University College of Medicine Seoul Korea
| | - Hanbyoul Cho
- Department of Obstetrics and Gynecology Yonsei University College of Medicine Seoul Korea
- Institute of Women's Life Medical Science Yonsei University College of Medicine Seoul Korea
| |
Collapse
|
15
|
Wang S, Yang C, Pan C, Feng X, Lei Z, Huang J, Wei X, Li F, Ma Y. Identification of key genes and functional enrichment pathways involved in fat deposition in Xinyang buffalo by WGCNA. Gene X 2022; 818:146225. [PMID: 35063576 DOI: 10.1016/j.gene.2022.146225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 12/06/2021] [Accepted: 01/13/2022] [Indexed: 01/02/2023] Open
Abstract
The Xinyang buffalo is a valuable and endangered domestic heritage resource in the Dabie Mountain region in China. With the increasing mechanization of agriculture, the Xinyang buffalo, mainly used for labor, faces unprecedented challenges. One of the feasible approaches to conserve and expand the species is to transfer Xinyang buffalo from service-use to meat-use, but the main hindrance to this transformation is the inferior meat quality of Xinyang buffalo, which is not popular with consumers. Based on the above, this study was conducted to evaluate the growth performance (n = 120) and slaughter performance (n = 3) of Xinyang buffalo and to measure the amino acid levels of the eye muscle (EM), and assess the meat quality. Later, transcriptome sequencing was performed on the subcutaneous fat of the back at six (n = 3) and 30 months of age (n = 3), together with the excavation of candidate genes associated with fat deposition using the weighted co-expression network analysis (WGCNA) method. The results showed that the slaughter rate of Xinyang buffalo was 43.09%, net meat percentage was 33.04%, the ocular area was 59.16 ± 7.58, the backfat thickness was 1.03 ± 0.16, and meat bone ratio was 3.29. The total amino acid contents were 0.63 g per gram of beef, which contained 0.05 g of essential amino acids, and the three most abundant amino acids were Ser (447.17 mg/g), Asp (29.8 mg/g), and Pro (27.24 mg/g). The WGCNA results showed that six phenotypes measured were significantly correlated with the turquoise module (r > 0.97, P < 0.001), and the genes in these modules were significantly enriched in the pathways related to substance metabolism and energy metabolisms, such as metabolic pathways, citrate cycle, and fatty acid metabolism. Meanwhile, six key candidate genes (FH, MECR, GPI, PANK3, ATP6V1A, PHYH) were identified, which were associated with growth and development, fat deposition, and intra-muscular amino acid levels (P < 0.05). In short, this study provides another feasible way to preserve buffalo and enriches the theory of its molecular genetic breeding.
Collapse
Affiliation(s)
- Shuzhe Wang
- Ningxia Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan, China; College of Life Sciences, Xinyang Normal University, Xinyang 464000, Henan, China
| | - Chaoyun Yang
- Ningxia Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan, China
| | - Cuili Pan
- Ningxia Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan, China
| | - Xue Feng
- Ningxia Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan, China
| | - Zhaoxiong Lei
- Ningxia Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan, China
| | - Jieping Huang
- Ningxia Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan, China; State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Xuefeng Wei
- Ningxia Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan, China; College of Life Sciences, Xinyang Normal University, Xinyang 464000, Henan, China
| | - Fen Li
- Ningxia Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan, China
| | - Yun Ma
- Ningxia Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan, China.
| |
Collapse
|
16
|
Pan S, Chen R. Pathological implication of protein post-translational modifications in cancer. Mol Aspects Med 2022; 86:101097. [PMID: 35400524 PMCID: PMC9378605 DOI: 10.1016/j.mam.2022.101097] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 03/21/2022] [Accepted: 03/22/2022] [Indexed: 02/07/2023]
Abstract
Protein post-translational modifications (PTMs) profoundly influence protein functions and play crucial roles in essentially all cell biological processes. The diverse realm of PTMs and their crosstalk is linked to many critical signaling events involved in neoplastic transformation, carcinogenesis and metastasis. The pathological roles of various PTMs are implicated in all aspects of cancer hallmark functions, cancer metabolism and regulation of tumor microenvironment. Study of PTMs has become an important area in cancer research to understand cancer biology and discover novel biomarkers and therapeutic targets. With a limited scope, this review attempts to discuss some PTMs of high frequency with recognized importance in cancer biology, including phosphorylation, acetylation, glycosylation, palmitoylation and ubiquitination, as well as their implications in clinical applications. These protein modifications are among the most abundant PTMs and profoundly implicated in carcinogenesis.
Collapse
|
17
|
Xia T, Chen D, Liu X, Qi H, Wang W, Chen H, Ling T, Otkur W, Zhang CS, Kim J, Lin SC, Piao HL. Midkine noncanonically suppresses AMPK activation through disrupting the LKB1-STRAD-Mo25 complex. Cell Death Dis 2022; 13:414. [PMID: 35487917 PMCID: PMC9054788 DOI: 10.1038/s41419-022-04801-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/23/2022] [Accepted: 03/30/2022] [Indexed: 11/09/2022]
Abstract
Midkine (MDK), a secreted growth factor, regulates signal transduction and cancer progression by interacting with receptors, and it can be internalized into the cytoplasm by endocytosis. However, its intracellular function and signaling regulation remain unclear. Here, we show that intracellular MDK interacts with LKB1 and STRAD to disrupt the LKB1-STRAD-Mo25 complex. Consequently, MDK decreases the activity of LKB1 to dampen both the basal and stress-induced activation of AMPK by glucose starvation or treatment of 2-DG. We also found that MDK accelerates cancer cell proliferation by inhibiting the activation of the LKB1-AMPK axis. In human cancers, compared to other well-known growth factors, MDK expression is most significantly upregulated in cancers, especially in liver, kidney and breast cancers, correlating with clinical outcomes and inversely correlating with phosphorylated AMPK levels. Our study elucidates an inhibitory mechanism for AMPK activation, which is mediated by the intracellular MDK through disrupting the LKB1-STRAD-Mo25 complex.
Collapse
|
18
|
Bibak B, Shakeri F, Keshavarzi Z, Mollazadeh H, Javid H, Jalili-Nik M, Sathyapalan T, Afshari AR, Sahebkar A. Anticancer mechanisms of Berberine: a good choice for glioblastoma multiforme therapy. Curr Med Chem 2022; 29:4507-4528. [PMID: 35209812 DOI: 10.2174/0929867329666220224112811] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 12/30/2021] [Accepted: 01/05/2022] [Indexed: 11/22/2022]
Abstract
The most typical malignant brain tumor, glioblastoma multiforme (GBM), seems to have a grim outcome, despite the intensive multi-modality interventions. Literature suggests that biologically active phytomolecules may exert anticancer properties by regulating several signaling pathways. Berberine, an isoquinoline alkaloid, has various pharmacological applications to combat severe diseases like cancer. Mechanistically, Berberine inhibits cell proliferation and invasion, suppresses tumor angiogenesis, and induces cell apoptosis. The effect of the antitumoral effect of Berberine in GBM is increasingly recognized. This review sheds new light on the regulatory signaling mechanisms of Berberine in various cancer, proposing its potential role as a therapeutic agent for GBM. .
Collapse
Affiliation(s)
- Bahram Bibak
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Farzaneh Shakeri
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Zakieh Keshavarzi
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Hamid Mollazadeh
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Hossein Javid
- Department of Medical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Jalili-Nik
- Department of Medical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Thozhukat Sathyapalan
- Academic Diabetes Endocrinology and Metabolism, Hull York Medical School, University of Hull, Hull, United Kingdom
| | - Amir R Afshari
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
19
|
Lau KH, Tan AM, Shi Y. New and Emerging Targeted Therapies for Advanced Breast Cancer. Int J Mol Sci 2022; 23:2288. [PMID: 35216405 PMCID: PMC8874375 DOI: 10.3390/ijms23042288] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/15/2022] [Accepted: 02/16/2022] [Indexed: 02/06/2023] Open
Abstract
In the United States, breast cancer is among the most frequently diagnosed cancers in women. Breast cancer is classified into four major subtypes: human epidermal growth factor receptor 2 (HER2), Luminal-A, Luminal-B, and Basal-like or triple-negative, based on histopathological criteria including the expression of hormone receptors (estrogen receptor and/or progesterone receptor) and/or HER2. Primary breast cancer treatments can include surgery, radiation therapy, systemic chemotherapy, endocrine therapy, and/or targeted therapy. Endocrine therapy has been shown to be effective in hormone receptor-positive breast cancers and is a common choice for adjuvant therapy. However, due to the aggressive nature of triple-negative breast cancer, targeted therapy is becoming a noteworthy area of research in the search for non-endocrine-targets in breast cancer. In addition to HER2-targeted therapy, other emerging therapies include immunotherapy and targeted therapy against critical checkpoints and/or pathways in cell growth. This review summarizes novel targeted breast cancer treatments and explores the possible implications of combination therapy.
Collapse
Affiliation(s)
| | | | - Yihui Shi
- Department of Basic Science, College of Medicine, California Northstate University, Elk Grove, CA 95757, USA; (K.H.L.); (A.M.T.)
| |
Collapse
|
20
|
Li T, Tian Y, Wang Y, Cui Z, He Z, Wu X, Zhang Y, Jiang H. Kiss1 Inhibits the Proliferation of Nasopharyngeal Carcinoma Cells Via Activation of the LKB1/AMPK Pathway. Front Oncol 2022; 11:724251. [PMID: 35117986 PMCID: PMC8804215 DOI: 10.3389/fonc.2021.724251] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 12/24/2021] [Indexed: 12/31/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a cancer that occurs in the nasopharynx. Infinite proliferation and distant metastasis are the main characteristics of NPC cells, and the main reason for the current failure of malignant tumor treatment. In this study, by integrating the immunohistochemical, cell transfection, western blot and real-time reverse transcriptase polymerase chain reaction (RT-PCR) analysis, we observed that the expression of KISS1 and its receptor gene (KISS1R) negatively related with the proliferation of NPC cells. Overexpression of the KISS1 genes in cells reduced cell proliferation, slow down the cell cycle, and increased apoptosis. Additionally, overexpression of these genes significantly increased Liver Kinase B1 (LKB1), phosphorylation of LKB1 and AMPK, indicated by Western blotting. Together, all of these results suggested for the first time that KISS1 and KISS1R suppress the proliferation of NPC cells by activating the LKB1/AMPK pathway, thus revealing a viable indicator for diagnosis of NPC in clinical practice.
Collapse
Affiliation(s)
- Tingting Li
- Department of Radiation Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yong Tian
- Department of Pharmacy, The First Affiliated Hospital of Bengbu Medical Collage, Bengbu, China
| | - Yixuan Wang
- General Surgery, Po Cheung Hospital, Bozhou, China
| | - Zhen Cui
- Department of Radiation Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Zelai He
- Department of Radiation Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Xiao Wu
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yajun Zhang
- Department of Radiation Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Hao Jiang
- Department of Radiation Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| |
Collapse
|
21
|
Dent P. Cell Signaling and Translational Developmental Therapeutics. COMPREHENSIVE PHARMACOLOGY 2022. [PMCID: PMC7538147 DOI: 10.1016/b978-0-12-820472-6.00002-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
The relationships between drug pharmacodynamics and subsequent changes in cellular signaling processes are complex. Many in vitro cell signaling studies often use drug concentrations above physiologically safe drug levels achievable in a patient's plasma. Drug companies develop agents to inhibit or modify the activities of specific target enzymes, often without a full consideration that their compounds have additional unknown targets. These two negative sequelae, when published together, become impediments against successful developmental therapeutics and translation because this data distorts our understanding of signaling mechanisms and reduces the probability of successfully translating drug-based concepts from the bench to the bedside. This article will discuss cellular signaling in isolation and as it relates to extant single and combined therapeutic drug interventions. This will lead to a hypothetical series standardized sequential approaches describing a rigorous concept to drug development and clinical translation.
Collapse
|
22
|
Chen Y, Maniakas A, Tan L, Cui M, Le X, Niedzielski JS, Michel KA, Harlan CJ, Lu W, Henderson YC, Mohamed ASR, Lorenzi PL, Putluri N, Bankson JA, Sandulache VC, Lai SY. Development of a rational strategy for integration of lactate dehydrogenase A suppression into therapeutic algorithms for head and neck cancer. Br J Cancer 2021; 124:1670-1679. [PMID: 33742144 PMCID: PMC8110762 DOI: 10.1038/s41416-021-01297-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 01/09/2021] [Accepted: 01/27/2021] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Lactate dehydrogenase (LDH) is a critical metabolic enzyme. LDH A (LDHA) overexpression is a hallmark of aggressive malignancies and has been linked to tumour initiation, reprogramming and progression in multiple tumour types. However, successful LDHA inhibition strategies have not materialised in the translational and clinical space. We sought to develop a rational strategy for LDHA suppression in the context of solid tumour treatment. METHODS We utilised a doxycycline-inducible short hairpin RNA (shRNA) system to generate LDHA suppression. Lactate and LDH activity levels were measured biochemically and kinetically using hyperpolarised 13C-pyruvate nuclear magnetic resonance spectroscopy. We evaluated effects of LDHA suppression on cellular proliferation and clonogenic survival, as well as on tumour growth, in orthotopic models of anaplastic thyroid carcinoma (ATC) and head and neck squamous cell carcinoma (HNSCC), alone or in combination with radiation. RESULTS shRNA suppression of LDHA generated a time-dependent decrease in LDH activity with transient shifts in intracellular lactate levels, a decrease in carbon flux from pyruvate into lactate and compensatory shifts in metabolic flux in glycolysis and the Krebs cycle. LDHA suppression decreased cellular proliferation and temporarily stunted tumour growth in ATC and HNSCC xenografts but did not by itself result in tumour cure, owing to the maintenance of residual viable cells. Only when chronic LDHA suppression was combined with radiation was a functional cure achieved. CONCLUSIONS Successful targeting of LDHA requires exquisite dose and temporal control without significant concomitant off-target toxicity. Combinatorial strategies with conventional radiation are feasible as long as the suppression is targeted, prolonged and non-toxic.
Collapse
Affiliation(s)
- Yunyun Chen
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anastasios Maniakas
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Hôpital Maisonneuve-Rosemont, University of Montreal, Montreal, QC, Canada
| | - Lin Tan
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Meng Cui
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Head Neck and Thyroid, Henan Cancer Hospital affiliated to Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Xiangdong Le
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joshua S Niedzielski
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Keith A Michel
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Collin J Harlan
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wuhao Lu
- Department of Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, TX, USA
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ying C Henderson
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Abdallah S R Mohamed
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Philip L Lorenzi
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nagireddy Putluri
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - James A Bankson
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vlad C Sandulache
- Department of Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, TX, USA.
| | - Stephen Y Lai
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
23
|
Wu Z, Xi P, Zhang Y, Wang H, Xue J, Sun X, Tian D. LKB1 up-regulation inhibits hypothalamic inflammation and attenuates diet-induced obesity in mice. Metabolism 2021; 116:154694. [PMID: 33358943 DOI: 10.1016/j.metabol.2020.154694] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 12/11/2020] [Accepted: 12/15/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Diet-induced obesity (DIO) is associated with chronic, low-grade inflammation in the hypothalamus. The inflammatory pathway of the hypothalamus is activated during obesity, and inhibition of activation of the inflammatory pathway can partially reverse obesity. Therefore, exploring new targets for inhibiting hypothalamic inflammation will provide new ideas for the prevention and treatment of obesity. Liver kinase B1 (LKB1), a serine/threonine kinase, is a tumor suppressor and metabolic regulator. Recent studies have shown that LKB1 has a certain anti-inflammatory effect. However, a role of LKB1 in the regulation of hypothalamic inflammation remains unclear. Therefore, we examined whether LKB1 overexpression in the hypothalamus could weaken the hypothalamic inflammation and inhibit the development of obesity. METHODS LKB1 overexpressing adeno-associated virus (AAV) particles were injected stereotactically into the third ventricle (3 V) of C57BL/6 mice fed with HFD. We assessed changes in body mass and adiposity, food intake, hypothalamic inflammatory markers, and energy and glucose metabolism. RESULTS LKB1 up-regulation in hypothalamus attenuated diet-induced hypothalamic inflammation, reduced food intake and body weight gain. In addition, the overexpression of hypothalamic LKB1 increased the insulin sensitivity and improved whole-body lipid metabolism, which attenuated hepatic fat accumulation and serum lipid levels. CONCLUSION Hypothalamic LKB1 up-regulation attenuates hypothalamic inflammation, and protects against hypothalamic inflammation induced damage to melanocortin system, resulting in lower food intake and lower fat mass accumulation, which consequently protects mice from the development of obesity. Our data suggest LKB1 as a novel negative regulator of hypothalamic inflammation, and also a potentially important target for treating other inflammatory diseases.
Collapse
Affiliation(s)
- Zhaoxia Wu
- Department of Clinical Laboratory Diagnostics, Tianjin Medical University, Tianjin 300203, China
| | - Pengjiao Xi
- Department of Clinical Laboratory Diagnostics, Tianjin Medical University, Tianjin 300203, China
| | - Yan Zhang
- Department of Clinical Laboratory Diagnostics, Tianjin Medical University, Tianjin 300203, China
| | - Haomin Wang
- Department of Human Anatomy and Histology, Tianjin Medical University, Tianjin 300070, China
| | - Jie Xue
- Department of Human Anatomy and Histology, Tianjin Medical University, Tianjin 300070, China
| | - Xuguo Sun
- Department of Clinical Laboratory Diagnostics, Tianjin Medical University, Tianjin 300203, China
| | - Derun Tian
- Department of Clinical Laboratory Diagnostics, Tianjin Medical University, Tianjin 300203, China; Department of Human Anatomy and Histology, Tianjin Medical University, Tianjin 300070, China.
| |
Collapse
|
24
|
Mobasher MA, Germoush MO, Galal El-Tantawi H, Samy El-Said K. Metformin Improves Biochemical and Pathophysiological Changes in Hepatocellular Carcinoma with Pre-Existed Diabetes Mellitus Rats. Pathogens 2021; 10:59. [PMID: 33440701 PMCID: PMC7830090 DOI: 10.3390/pathogens10010059] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/24/2020] [Accepted: 12/28/2020] [Indexed: 12/13/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the world's most widely recognized malignant tumors that accounts for 90% of all the primary liver cancers and is a major cause of death from cancer, representing half a million deaths per year. Obesity and associated metabolic irregularities, particularly diabetes mellitus (DM) and insulin resistance, are important risk factors for the advancement of HCC. Recently, retrospective studies showed that metformin (MET) could protect the hepatic tissues in pre-existing diabetes mellitus from HCC. The purpose of this study was to assess the role of MET treatment in the pre-existing diabetic rats before and after HCC induction by diethylnitrosamine (DEN). Thirty-five male Sprague Dawley albino rats were partitioned into the following groups: Group 1 (Gp1) was the control. Gp2 was injected intraperitoneally (i.p) with streptozotocin (STZ) (80 mg/kg) and DEN (50 mg/kg/7 weeks). Gp3, Gp4, and Gp5 were injected as in Gp2 and treated with MET (150 mg/kg) before and/or after HCC induction. Biochemical parameters including liver functions, lipid profile, and oxidative stress biomarkers were determined. Furthermore, histological and immunohistochemical changes were assessed in all groups. Our results illustrated that the group of rats that were treated with STZ and DEN had significant changes in both liver functions and were associated with alterations in the liver histopathological architectures. Treatment with MET before or after HCC induction ameliorated the cellular changes in the liver tissues; however, the utmost protection was found in a group of rats, which were treated with MET before and after HCC induction.
Collapse
Affiliation(s)
- Maysa A. Mobasher
- Department of Pathology, Biochemistry Division, College of Medicine, Jouf University, Sakaka 41412, Saudi Arabia
- Department of Clinical Pathology, El Ahrar Educational Hospital, Ministry of Health, Zagazig 44511, Egypt
| | - Mousa O. Germoush
- Biology Department, College of Science, Jouf University, Sakaka 41412, Saudi Arabia;
| | | | - Karim Samy El-Said
- Chemistry Department, Biochemistry Division, Faculty of Science, Tanta University, Tanta 31527, Egypt;
| |
Collapse
|
25
|
Tyszka-Czochara M. Caffeic Acid targets metabolism of cervical squamous cell carcinoma. Cancer 2021. [DOI: 10.1016/b978-0-12-819547-5.00025-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
26
|
Babaei G, Aziz SGG, Jaghi NZZ. EMT, cancer stem cells and autophagy; The three main axes of metastasis. Biomed Pharmacother 2020; 133:110909. [PMID: 33227701 DOI: 10.1016/j.biopha.2020.110909] [Citation(s) in RCA: 296] [Impact Index Per Article: 59.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/29/2020] [Accepted: 10/17/2020] [Indexed: 02/07/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) and Cancer stem-like cells (CSCs) are major factors contributing to the metastasis of cancer cells. Consequently, the signaling pathways involved in both processes are appropriate therapeutic targets in the treatment of metastasis. Autophagy is another process that has recently attracted the attention of many researchers; depending on the type of cancer and tissue and the stage of cancer, this process can play a dual role in the development of cancer cells. Studies on cancer cells have shown that different signaling pathways are involved in all three processes, namely, cancer stem cells, autophagy, and EMT. The purpose of this study was to investigate and elucidate the relationship between the effective signaling pathways in all three processes, which could play an effective role in determining appropriate therapeutic goals.
Collapse
Affiliation(s)
- Ghader Babaei
- Department of Biochemistry, Faculty of Medicine, Urmia University Medical Sciences (UMSU), Urmia, Iran; Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran.
| | | | - Nasrin Zare Zavieyh Jaghi
- Department of Biochemistry, Faculty of Medicine, Urmia University Medical Sciences (UMSU), Urmia, Iran
| |
Collapse
|
27
|
Zhang M, Liang L, He J, He Z, Yue C, Jin X, Gao M, Xiao S, Zhou Y. Fra-1 Inhibits Cell Growth and the Warburg Effect in Cervical Cancer Cells via STAT1 Regulation of the p53 Signaling Pathway. Front Cell Dev Biol 2020; 8:579629. [PMID: 33102485 PMCID: PMC7554318 DOI: 10.3389/fcell.2020.579629] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 09/08/2020] [Indexed: 12/31/2022] Open
Abstract
The oncogenesis of cervical cancer is a multi-factor and multi-step process, and major risk factors include oncogene activation with tumor suppressor gene inactivation, viral factors, and immune factors. For example, the human papillomavirus (HPV) has been linked to the occurrence of cervical cancer. At present, the pathogenesis of cervical cancer remains unclear. Fra-1 (Fos-related antigen 1, also known as FOSL1) is a member of the Fos family and an important nuclear transcription factor that regulates normal cell growth, differentiation, and apoptosis. In the present study, we found that Fra-1 inhibited the proliferation of cervical cancer cells while also promoting apoptosis and affecting cell cycle distribution. Moreover, Fra-1 up-regulated STAT1 expression and modulated p53 signal pathway activity in cervical cancer cells. Overexpression of Fra-1 inhibited cell senescence by altering sirtuin 1 (SIRT1) expression in HeLa cells, and Fra-1 overexpression restored mitochondrial disorder and suppressed metabolic reprogramming in HeLa cells. Silencing of STAT1 impaired the inhibitory effect of Fra-1 on cervical cancer cell growth, while knock-down of STAT1 reversed the effect on cell senescence and mitochondrial dysfunction caused by Fra-1 in HeLa cells. Silencing of STAT1 also recovered metabolic reprogramming in cervical cancer cells. In summary, our results show that Fra-1 inhibited cervical cancer cell growth and the Warburg effect via STAT1-mediated regulation of the p53 signaling pathway.
Collapse
Affiliation(s)
- Manying Zhang
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Lin Liang
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Junyu He
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Zhengxi He
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Chunxue Yue
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Xi Jin
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Mengxiang Gao
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Songshu Xiao
- Department of Gynecology and Obstetrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yanhong Zhou
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
28
|
Gao N, Yang F, Chen S, Wan H, Zhao X, Dong H. The role of TRPV1 ion channels in the suppression of gastric cancer development. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:206. [PMID: 33008449 PMCID: PMC7531167 DOI: 10.1186/s13046-020-01707-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 09/10/2020] [Indexed: 12/19/2022]
Abstract
Background Although the aberrant expression and function of most Ca2+-permeable channels are known to promote gastrointestinal tumors, the association between transient receptor potential vanilloid receptor 1 (TRPV1) channels and gastric cancer (GC) has not yet been explored. Herein, we sought to determine the role of TRPV1 channels in the development of GC and to elucidate the underlying molecular mechanisms involved therein. Methods Immunohistochemistry, qPCR, Western blot, immunofluorescence assays were used to detect the mRNA and protein expression of TRPV1 in GC cells and tissues, and the clinical significance of TRPV1 in GC was also studied by clinicopathologic analysis. CCK8, colony formation, flow cytometry assays were used to detect the proliferation and survival of GC cells, while transwell assay was used to detect migration and invasion of GC cells in vitro. Tumor xenograft and peritoneal dissemination assays in nude mice were used to examine the role of TRPV1 in GC development in vivo. Results TRPV1 expression was significantly downregulated in human primary GC tissues compared to their adjacent tissues. The decreased expression of TRPV1 proteins in GC tissues was positively correlated with tumor size, histological grade, lymphatic metastasis, clinical stage, and was strongly correlated with poor prognosis of GC patients. Moreover, the expression of TRPV1 was closely correlated with Ki67, VEGFR, and E-cadherin, all of which are the well-known cancer markers for proliferation and metastasis. TRPV1 proteins were predominately expressed on the plasma membrane in several GC cell lines. TRPV1 overexpression blocked cell cycle at G1 phase to inhibit GC cell proliferation and attenuated migration and invasion of GC cells in vitro, but TRPV1 knockdown increased these parameters. TRPV1 significantly reduced gastric tumor size, number and peritoneal dissemination in vivo. Mechanistically, TRPV1 overexpression in GC cells increased [Ca2+]i, activated CaMKKβ and AMPK phosphorylation, and decreased expression of cyclin D1 and MMP2, while TRPV1 knockdown induced the opposite effects. Conclusions TRPV1 uniquely suppresses GC development through a novel Ca2+/CaMKKβ/AMPK pathway and its downregulation is correlated with poor survival of human GC patients. Thus, TRPV1 upregulation and its downstream signaling may represent a promising target for GC prevention and therapy.
Collapse
Affiliation(s)
- Nannan Gao
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Feng Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Siyuan Chen
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Hanxing Wan
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Xiaoyan Zhao
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
| | - Hui Dong
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China. .,Department of Medicine, School of Medicine, University of California, San Diego, CA, 92093, USA.
| |
Collapse
|
29
|
Figueroa-González G, Carrillo-Hernández JF, Perez-Rodriguez I, Cantú de León D, Campos-Parra AD, Martínez-Gutiérrez AD, Coronel-Hernández J, García-Castillo V, López-Camarillo C, Peralta-Zaragoza O, Jacobo-Herrera NJ, Guardado-Estrada M, Pérez-Plasencia C. Negative Regulation of Serine Threonine Kinase 11 (STK11) through miR-100 in Head and Neck Cancer. Genes (Basel) 2020; 11:1058. [PMID: 32911741 PMCID: PMC7563199 DOI: 10.3390/genes11091058] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/18/2020] [Accepted: 08/25/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Serine Threonine Kinase 11 (STK11), also known as LKB1, is a tumor suppressor gene that regulates several biological processes such as apoptosis, energetic metabolism, proliferation, invasion, and migration. During malignant progression, different types of cancer inhibit STK11 function by mutation or epigenetic inactivation. In Head and Neck Cancer, it is unclear what mechanism is involved in decreasing STK11 levels. Thus, the present work aims to determine whether STK11 expression might be regulated through epigenetic or post-translational mechanisms. METHODS Expression levels and methylation status for STK11 were analyzed in 59 cases of head and neck cancer and 10 healthy tissue counterparts. Afterward, we sought to identify candidate miRNAs exerting post-transcriptional regulation of STK11. Then, we assessed a luciferase gene reporter assay to know if miRNAs directly target STK11 mRNA. The expression levels of the clinical significance of mir-100-3p, -5p, and STK11 in 495 HNC specimens obtained from the TCGA database were further analyzed. Finally, the Kaplan-Meier method was used to estimate the prognostic significance of the miRNAs for Overall Survival, and survival curves were compared through the log-rank test. RESULTS STK11 was under-expressed, and its promoter region was demethylated or partially methylated. miR-17-5p, miR-106a-5p, miR-100-3p, and miR-100-5p could be negative regulators of STK11. Our experimental data suggested evidence that miR-100-3p and -5p were over-expressed in analyzed tumor patient samples. Luciferase gene reporter assay experiments showed that miR-100-3p targets and down-regulates STK11 mRNA directly. With respect to overall survival, STK11 expression level was significant for predicting clinical outcomes. CONCLUSION This is, to our knowledge, the first report of miR-100-3p targeting STK11 in HNC. Together, these findings may support the importance of regulation of STK11 through post-transcriptional regulation in HNC and the possible contribution to the carcinogenesis process in this neoplasia.
Collapse
Affiliation(s)
- Gabriela Figueroa-González
- Unidad Multidisciplinaria de Investigación Experimental Zaragoza (UMIEZ), Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Mexico City 09230, Mexico;
- Unidad de Investigación Biomédica en Cáncer, Laboratorio de Genómica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico; (J.F.C.-H.); (I.P.-R.); (D.C.d.L.); (A.D.C.-P.); (A.D.M.-G.); (J.C.-H.)
| | - José F. Carrillo-Hernández
- Unidad de Investigación Biomédica en Cáncer, Laboratorio de Genómica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico; (J.F.C.-H.); (I.P.-R.); (D.C.d.L.); (A.D.C.-P.); (A.D.M.-G.); (J.C.-H.)
| | - Itzel Perez-Rodriguez
- Unidad de Investigación Biomédica en Cáncer, Laboratorio de Genómica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico; (J.F.C.-H.); (I.P.-R.); (D.C.d.L.); (A.D.C.-P.); (A.D.M.-G.); (J.C.-H.)
| | - David Cantú de León
- Unidad de Investigación Biomédica en Cáncer, Laboratorio de Genómica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico; (J.F.C.-H.); (I.P.-R.); (D.C.d.L.); (A.D.C.-P.); (A.D.M.-G.); (J.C.-H.)
| | - Alma D. Campos-Parra
- Unidad de Investigación Biomédica en Cáncer, Laboratorio de Genómica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico; (J.F.C.-H.); (I.P.-R.); (D.C.d.L.); (A.D.C.-P.); (A.D.M.-G.); (J.C.-H.)
| | - Antonio D. Martínez-Gutiérrez
- Unidad de Investigación Biomédica en Cáncer, Laboratorio de Genómica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico; (J.F.C.-H.); (I.P.-R.); (D.C.d.L.); (A.D.C.-P.); (A.D.M.-G.); (J.C.-H.)
| | - Jossimar Coronel-Hernández
- Unidad de Investigación Biomédica en Cáncer, Laboratorio de Genómica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico; (J.F.C.-H.); (I.P.-R.); (D.C.d.L.); (A.D.C.-P.); (A.D.M.-G.); (J.C.-H.)
| | - Verónica García-Castillo
- Unidad de Investigación Biomédica en Cáncer, Laboratorio de Genómica del Cáncer, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Edo.Mex, Mexico;
| | - César López-Camarillo
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, Mexico City 09790, Mexico;
| | - Oscar Peralta-Zaragoza
- Dirección de Infecciones Crónicas y Cáncer, Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca 62100, Morelos, Mexico;
| | - Nadia J. Jacobo-Herrera
- Unidad de Bioquímica, Instituto Nacional de Nutrición y Ciencias Médicas, Salvador Zubirán, Mexico City 14000, Mexico;
| | - Mariano Guardado-Estrada
- Laboratorio de Genética, Licenciatura en Ciencia Forense, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04360, Mexico;
| | - Carlos Pérez-Plasencia
- Unidad de Investigación Biomédica en Cáncer, Laboratorio de Genómica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico; (J.F.C.-H.); (I.P.-R.); (D.C.d.L.); (A.D.C.-P.); (A.D.M.-G.); (J.C.-H.)
- Unidad de Investigación Biomédica en Cáncer, Laboratorio de Genómica del Cáncer, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Edo.Mex, Mexico;
| |
Collapse
|
30
|
Samec M, Liskova A, Koklesova L, Samuel SM, Zhai K, Buhrmann C, Varghese E, Abotaleb M, Qaradakhi T, Zulli A, Kello M, Mojzis J, Zubor P, Kwon TK, Shakibaei M, Büsselberg D, Sarria GR, Golubnitschaja O, Kubatka P. Flavonoids against the Warburg phenotype-concepts of predictive, preventive and personalised medicine to cut the Gordian knot of cancer cell metabolism. EPMA J 2020; 11:377-398. [PMID: 32843908 PMCID: PMC7429635 DOI: 10.1007/s13167-020-00217-y] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 06/30/2020] [Indexed: 01/10/2023]
Abstract
The Warburg effect is characterised by increased glucose uptake and lactate secretion in cancer cells resulting from metabolic transformation in tumour tissue. The corresponding molecular pathways switch from oxidative phosphorylation to aerobic glycolysis, due to changes in glucose degradation mechanisms known as the 'Warburg reprogramming' of cancer cells. Key glycolytic enzymes, glucose transporters and transcription factors involved in the Warburg transformation are frequently dysregulated during carcinogenesis considered as promising diagnostic and prognostic markers as well as treatment targets. Flavonoids are molecules with pleiotropic activities. The metabolism-regulating anticancer effects of flavonoids are broadly demonstrated in preclinical studies. Flavonoids modulate key pathways involved in the Warburg phenotype including but not limited to PKM2, HK2, GLUT1 and HIF-1. The corresponding molecular mechanisms and clinical relevance of 'anti-Warburg' effects of flavonoids are discussed in this review article. The most prominent examples are provided for the potential application of targeted 'anti-Warburg' measures in cancer management. Individualised profiling and patient stratification are presented as powerful tools for implementing targeted 'anti-Warburg' measures in the context of predictive, preventive and personalised medicine.
Collapse
Affiliation(s)
- Marek Samec
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Alena Liskova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Lenka Koklesova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine in Qatar, Education City, Qatar Foundation, 24144, Doha, Qatar
| | - Kevin Zhai
- Department of Physiology and Biophysics, Weill Cornell Medicine in Qatar, Education City, Qatar Foundation, 24144, Doha, Qatar
| | - Constanze Buhrmann
- Musculoskeletal Research Group and Tumour Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, 80336 Munich, Germany
| | - Elizabeth Varghese
- Department of Physiology and Biophysics, Weill Cornell Medicine in Qatar, Education City, Qatar Foundation, 24144, Doha, Qatar
| | - Mariam Abotaleb
- Department of Physiology and Biophysics, Weill Cornell Medicine in Qatar, Education City, Qatar Foundation, 24144, Doha, Qatar
| | - Tawar Qaradakhi
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3011 Australia
| | - Anthony Zulli
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3011 Australia
| | - Martin Kello
- Department of Pharmacology, Faculty of Medicine, P. J. Šafarik University, 040 11 Košice, Slovakia
| | - Jan Mojzis
- Department of Pharmacology, Faculty of Medicine, P. J. Šafarik University, 040 11 Košice, Slovakia
| | - Pavol Zubor
- Department of Gynecologic Oncology, Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
- OBGY Health & Care, Ltd., 01001 Zilina, Slovak Republic
| | - Taeg Kyu Kwon
- Department of Immunology and School of Medicine, Keimyung University, Dalseo-Gu, Daegu, 426 01 South Korea
| | - Mehdi Shakibaei
- Musculoskeletal Research Group and Tumour Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, 80336 Munich, Germany
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine in Qatar, Education City, Qatar Foundation, 24144, Doha, Qatar
| | - Gustavo R. Sarria
- Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Olga Golubnitschaja
- Predictive, Preventive Personalised (3P) Medicine, Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia
| |
Collapse
|
31
|
Luteolin Protects Against CIRI, Potentially via Regulation of the SIRT3/AMPK/mTOR Signaling Pathway. Neurochem Res 2020; 45:2499-2515. [PMID: 32809175 DOI: 10.1007/s11064-020-03108-w] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 06/28/2020] [Accepted: 08/01/2020] [Indexed: 12/21/2022]
Abstract
Mitochondrial abnormalities accelerate the progression of ischemic brain damage. Sirtuin 3 (SIRT3) is mainly found in mitochondria and affects almost all major aspects of mitochondrial function. Luteolin, a flavonoid with diverse biological properties, including antioxidant activity, inhibition of cell apoptosis and regulation of autophagy. It also modulates the activity of AMP activated kinase and/or sirtuin 1 (SIRT 1) by regulating the expression of sirtuins. We investigated the protective effects of luteolin on cerebral ischemia-reperfusion. It was found through experiments that luteolin reduced the infarcted area of MCAO rat model, and based on the experimental results, it was inferred that luteolin affected the AMPK, mTOR and SIRT3 pathways, thereby protecting brain cells. As expected, we found that luteolin can reduce the neurological function score, the degree of cerebral edema, the cerebral infarction volume, alleviate morphological changes in the cortex and hippocampus, increase neuron survival and decrease the number of apoptotic neurons. At the same time, luteolin significantly reduced the number of GFAP and Iba-1 positive glial cells in the hippocampus while enhanced the scavenging of oxygen free radicals and the activity of SOD in mitochondria. Addtionally, it can also enhance antioxidant capacity via the reversal of mitochondrial swelling and the mitochondrial transmembrane potential. Moreover, luteolin can increase SIRT3-targeted expression in mitochondria, decrease the phosphorylation of AMPK, and increase phosphor-mTOR (p-mTOR) levels, which may have occurred specifically through activation of the SIRT3/AMPK/mTOR pathway. We speculate that luteolin reduces the pathological progression of CIRI by increasing SIRT3 expression and enhancing mitochondrial function. Therefore, the results indicate that luteolin can increase the transduction of SIRT3, providing a potential mechanism for neuroprotective effects in patients with cerebral ischemia.
Collapse
|
32
|
Wu Z, Han J, Xue J, Xi P, Wang H, He L, Wang Q, Liang H, Sun X, Tian D. Deletion of liver kinase B1 in POMC neurons predisposes to diet-induced obesity. Life Sci 2020; 258:118204. [PMID: 32763296 DOI: 10.1016/j.lfs.2020.118204] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/27/2020] [Accepted: 08/01/2020] [Indexed: 01/10/2023]
Abstract
AIMS Liver kinase B1 (LKB1) is a serine/threonine kinase. Although many biological functions of LKB1 have been identified, the role of hypothalamic LKB1 in the regulation of central energy metabolism and susceptibility to obesity is unknown. Therefore, we constructed POMC neuron-specific LKB1 knockout mice (PomcLkb1 KO) and studied it at the physiological, morphological, and molecular biology levels. MAIN METHODS Eight-week-old male PomcLkb1 KO mice and their littermates were fed a standard chow fat diet (CFD) or a high-fat diet (HFD) for 3 months. Body weight and food intake were monitored. Dual-energy X-ray absorptiometry was used to measure the fat mass and lean mass. Glucose and insulin tolerance tests and serum biochemical markers were evaluated in the experimental mice. In addition, the levels of peripheral lipogenesis genes and central energy metabolism were measured. KEY FINDINGS PomcLkb1 KO mice did not exhibit impairments under normal physiological conditions. After HFD intervention, the metabolic phenotype of the PomcLkb1 KO mice changed, manifesting as increased food intake and an enhanced obesity phenotype. More seriously, PomcLkb1 KO mice showed increased leptin resistance, worsened hypothalamic inflammation and reduced POMC neuronal expression. SIGNIFICANCE We provide evidence that LKB1 in POMC neurons plays a significant role in regulating energy homeostasis. LKB1 in POMC neurons emerges as a target for therapeutic intervention against HFD-induced obesity and metabolic diseases.
Collapse
Affiliation(s)
- Zhaoxia Wu
- Department of Clinical Laboratory Diagnostics, Tianjin Medical University, Tianjin 300203, China
| | - Jie Han
- Department of Human Anatomy and Histology, Tianjin Medical University, Tianjin 300070, China
| | - Jie Xue
- Department of Human Anatomy and Histology, Tianjin Medical University, Tianjin 300070, China
| | - Pengjiao Xi
- Department of Clinical Laboratory Diagnostics, Tianjin Medical University, Tianjin 300203, China
| | - Haomin Wang
- Department of Human Anatomy and Histology, Tianjin Medical University, Tianjin 300070, China
| | - Lu He
- Department of Human Anatomy and Histology, Tianjin Medical University, Tianjin 300070, China
| | - Qiming Wang
- Department of Human Anatomy and Histology, Tianjin Medical University, Tianjin 300070, China
| | - Huimin Liang
- Department of School of Nursing, Tianjin Medical University, Tianjin 300070, China
| | - Xuguo Sun
- Department of Clinical Laboratory Diagnostics, Tianjin Medical University, Tianjin 300203, China.
| | - Derun Tian
- Department of Clinical Laboratory Diagnostics, Tianjin Medical University, Tianjin 300203, China; Department of Human Anatomy and Histology, Tianjin Medical University, Tianjin 300070, China.
| |
Collapse
|
33
|
Sarfraz I, Rasul A, Hussain G, Shah MA, Zahoor AF, Asrar M, Selamoglu Z, Ji XY, Adem Ş, Sarker SD. 6-Phosphogluconate dehydrogenase fuels multiple aspects of cancer cells: From cancer initiation to metastasis and chemoresistance. Biofactors 2020; 46:550-562. [PMID: 32039535 DOI: 10.1002/biof.1624] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 01/21/2020] [Indexed: 12/19/2022]
Abstract
Reprogrammed metabolism is key biochemical characteristic of malignant cells, which represents one of the emerging hallmarks of cancer. Currently, there is rising contemplation on oxidative pentose phosphate pathway (PPP) enzymes as potential therapeutic hits due to their affiliation with tumor metabolism. 6-Phosphogluconate dehydrogenase (6PGD), third oxidative decarboxylase of PPP, has received a great deal of attention during recent years due to its critical role in tumorigenesis and redox homeostasis. 6PGD has been reported to overexpress in number of cancer types and its hyperactivation is mediated through post-transcriptional and post-translational modifications by YTH domain family 2 (YTHDF2), Nrf2 (nuclear factor erythroid 2-related factor 2), EGFR (epidermal growth factor receptor) and via direct structural interactions with ME1 (malic enzyme 1). Upregulated expression of 6PGD provides metabolic as well as defensive advantage to cancer cells, thus, promoting their proliferative and metastatic potential. Moreover, enhanced 6PGD expression also performs key role in development of chemoresistance as well as radiation resistance in cancer. This review aims to discuss the historical timeline and cancer-specific role of 6PGD, pharmacological and genetic inhibitors of 6PGD and 6PGD as prognostic biomarker in order to explore its potential for therapeutic interventions. We anticipate that targeting this imperative supplier of NADPH might serve as tempting avenue to combat the deadly disease like cancer.
Collapse
Affiliation(s)
- Iqra Sarfraz
- Department of Zoology, Faculty of Life Sciences, Government College University Faisalabad (GCUF), Faisalabad, Pakistan
| | - Azhar Rasul
- Department of Zoology, Faculty of Life Sciences, Government College University Faisalabad (GCUF), Faisalabad, Pakistan
| | - Ghulam Hussain
- Neurochemical Biology and Genetics Laboratory (NGL), Department of Physiology, Faculty of Life Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Muhammad Ajmal Shah
- Department of Pharmacognosy, Faculty of Pharmaceutical Sciences, Government College University Faisalabad (GCUF), Faisalabad, Pakistan
| | - Ameer Fawad Zahoor
- Department of Chemistry, Faculty of Physical Sciences, Government College University Faisalabad (GCUF), Faisalabad, Pakistan
| | - Muhammad Asrar
- Department of Zoology, Faculty of Life Sciences, Government College University Faisalabad (GCUF), Faisalabad, Pakistan
| | - Zeliha Selamoglu
- Department of Medical Biology, Faculty of Medicine, Nigde Ömer Halisdemir University, Nigde, Turkey
| | - Xin-Ying Ji
- Henan International Joint Laboratory of Nuclear Protein Regulation, College of Medicine, Henan University, Kaifeng, China
| | - Şevki Adem
- Department of Chemistry, Faculty of Sciences, Çankırı Karatekin University, Çankırı, Turkey
| | - Satyajit D Sarker
- School of Pharmacy & Biomolecular Sciences, Liverpool John Moores University, London, UK
| |
Collapse
|
34
|
Li Z, Xu J, Cui H, Song J, Chen J, Wei J. Bioinformatics analysis of key biomarkers and potential molecular mechanisms in hepatocellular carcinoma induced by hepatitis B virus. Medicine (Baltimore) 2020; 99:e20302. [PMID: 32443377 PMCID: PMC7254842 DOI: 10.1097/md.0000000000020302] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) accounts for up to 90% of all primary hepatic malignancies; it is the sixth most common cancer and the second most common cause of cancer mortality worldwide. Numerous studies have shown that hepatitis B virus and its products, HBV integration, and mutation can induce HCC. However, the molecular mechanisms underpinning the regulation of HCC induced by HBV remain unclear. METHODS We downloaded 2 gene expression profiling datasets, of HBV and of HCC induced by HBV, from the gene expression omnibus (GEO) database. Differentially expressed genes (DEGs) between HCC and HBV were identified to explore any predisposing changes in gene expression associated with HCC. DEGs between HCC and adjacent healthy tissues were investigated to identify genes that may play a key role in HCC. Any overlapping genes among these DEGs were included in our bioinformatics analysis. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses of overlapping genes were performed using the Metascape online database; the protein-protein interaction (PPI) network was analyzed using the STRING online database; and we obtained the hub genes of the PPI network using Cytoscape software. An overall survival (OS) analysis of hub genes was performed using km-plotter and the gene expression profiling interactive analysis (GEPIA) online database. The expression levels of hub genes were determined using the TCGA and GEPIA databases. Finally, the relationships between hub genes and tumors were analyzed using the comparative toxicogenomics database (CTD). RESULTS We identified 113 overlapping genes from the 2 datasets. Using functional and pathway analyses, we found that the overlapping genes were mainly related to the AMPK signaling pathway and cellular responses to cadmium ions. C8A, SPP2, KLKB1, PROZ, C6, FETUB, MBL2, HGFAC, C8B, and ANGPTL3 were identified as hub genes and C8A, SPP2, PROZ, C6, HGFAC, and C8B were found to be significant for survival. CONCLUSION The DEGs re-analyzed between HCC and hepatitis B enable a systematic understanding of the molecular mechanisms of HCC reliant on hepatitis B virus.
Collapse
|
35
|
Jung M, Lee JH, Lee C, Park JH, Park YR, Moon KC. Prognostic Implication of pAMPK Immunohistochemical Staining by Subcellular Location and Its Association with SMAD Protein Expression in Clear Cell Renal Cell Carcinoma. Cancers (Basel) 2019; 11:cancers11101602. [PMID: 31640193 PMCID: PMC6826619 DOI: 10.3390/cancers11101602] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/17/2019] [Accepted: 10/19/2019] [Indexed: 11/18/2022] Open
Abstract
Although cytoplasmic AMP-activated protein kinase (AMPK) has been known as a tumor-suppressor protein, nuclear AMPK is suggested to support clear cell renal cell carcinoma (ccRCC). In addition, pAMPK interacts with TGF-β/SMAD, which is one of the frequently altered pathways in ccRCC. In this study, we investigated the prognostic significance of pAMPK with respect to subcellular location and investigated its interaction with TGF-β/SMAD in ccRCC. Immunohistochemical staining for pAMPK, pSMAD2 and SMAD4 was conducted on tissue microarray of 987 ccRCC specimens. Moreover, the levels of pSMAD2 were measured in Caki-1 cells treated with 5-aminoimidazole-4-carboxamide ribonucleotide. The relationship between AMPK/pAMPK and TGFB1 expression was determined using the TCGA database. As a result, pAMPK positivity, either in the cytoplasm or nuclei, was independently associated with improved ccRCC prognosis, after adjusting for TNM stage and WHO grade. Furthermore, pAMPK-positive ccRCC displayed increased pSMAD2 and SMAD4 expression, while activation of pAMPK increased pSMAD2 in Caki-1 cells. However, AMPK/pAMPK expression was inversely correlated with TGFB1 expression in the TCGA database. Therefore, pAMPK immunostaining, both in the cytoplasm and nuclei, is a useful prognostic biomarker for ccRCC. pAMPK targets TGF-β-independent phosphorylation of SMAD2 and activates pSMAD2/SMAD4, representing a novel anti-tumoral mechanism of pAMPK in ccRCC.
Collapse
Affiliation(s)
- Minsun Jung
- Department of Pathology, Seoul National University Hospital, Seoul 03080, Korea.
| | - Jeong Hoon Lee
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul 03722, Korea.
| | - Cheol Lee
- Department of Pathology, Seoul National University Hospital, Seoul 03080, Korea.
| | - Jeong Hwan Park
- Department of Pathology, SMG-SNU Boramae Medical Center, Seoul 07061, Korea.
| | - Yu Rang Park
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul 03722, Korea.
| | - Kyung Chul Moon
- Department of Pathology, Seoul National University Hospital, Seoul 03080, Korea.
- Kidney Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul 03080, Korea.
| |
Collapse
|
36
|
Liu J, Li X, Lu Q, Ren D, Sun X, Rousselle T, Li J, Leng J. AMPK: a balancer of the renin-angiotensin system. Biosci Rep 2019; 39:BSR20181994. [PMID: 31413168 PMCID: PMC6722492 DOI: 10.1042/bsr20181994] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 07/24/2019] [Accepted: 08/12/2019] [Indexed: 02/07/2023] Open
Abstract
The renin-angiotensin system (RAS) is undisputedly well-studied as one of the oldest and most critical regulators for arterial blood pressure, fluid volume, as well as renal function. In recent studies, RAS has also been implicated in the development of obesity, diabetes, hyperlipidemia, and other diseases, and also involved in the regulation of several signaling pathways such as proliferation, apoptosis and autophagy, and insulin resistance. AMP-activated protein kinase (AMPK), an essential cellular energy sensor, has also been discovered to be involved in these diseases and cellular pathways. This would imply a connection between the RAS and AMPK. Therefore, this review serves to draw attention to the cross-talk between RAS and AMPK, then summering the most recent literature which highlights AMPK as a point of balance between physiological and pathological functions of the RAS.
Collapse
Affiliation(s)
- Jia Liu
- Department of Geriatrics, The First Hospital of Jilin University, Changchun 130021, China
- Department of Surgery, University of South Florida, Tampa, FL 33612, U.S.A
| | - Xuan Li
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, U.S.A
| | - Qingguo Lu
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, U.S.A
| | - Di Ren
- Department of Surgery, University of South Florida, Tampa, FL 33612, U.S.A
| | - Xiaodong Sun
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, U.S.A
| | - Thomas Rousselle
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, U.S.A
| | - Ji Li
- Department of Surgery, University of South Florida, Tampa, FL 33612, U.S.A
| | - Jiyan Leng
- Department of Geriatrics, The First Hospital of Jilin University, Changchun 130021, China
| |
Collapse
|
37
|
Shen Y, Zhou N, An J, Zhang J, Wang M, Li Y, Jiang P. Haemophilus parasuis infection in 3D4/21 cells induces autophagy through the AMPK pathway. Cell Microbiol 2019; 21:e13031. [PMID: 30977277 DOI: 10.1111/cmi.13031] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 03/27/2019] [Accepted: 04/08/2019] [Indexed: 12/19/2022]
Abstract
Haemophilus parasuis (H. parasuis) is a common commensal in the upper respiratory tract of pigs, but causes Glässer's disease in stress conditions. To date, many studies focused on the immune evasion and virulence of H. parasuis; very few have focused on the role autophagy played in H. parasuis infection, particularly in porcine alveolar macrophages (PAMs). In this study, a PAM cell line, 3D4/21 cells were used to study the role of autophagy in H. parasuis infection. 3D4/21 cells tandemly expressing GFP, mCherry, and LC3 were infected with H. parasuis serovar 5 (Hps5). Western blot analysis and confocal and transmission electron microscopy showed that H. parasuis infection effectively induces autophagy. Using Hps strains of varying virulence (Hps4, Hps5, and Hps7) and UV-inactivated Hps5, we demonstrated that autophagy is associated with the internalisation of living virulent strains into cells. In 3D4/21 cells pretreated with rapamycin and 3-MA then infected by Hps4, Hps5, and Hps7, we demonstrated that autophagy affects invasion of H. parasuis in cells. AMPK signal results showed that Hps5 infection can upregulate the phosphorylation level of AMPK, which is consistent with the autophagy development. 3D4/21 cells pretreated with AICAR or Compound C then infected by Hps5 revealed that the autophagy induced by Hps5 infection is associated with the AMPK pathway. Our study contributes to the theoretical basis for the study of H. parasuis pathogenesis and development of novel drugs target for prevention Glässer's disease.
Collapse
Affiliation(s)
- Yijuan Shen
- Key Laboratory of Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Nini Zhou
- Key Laboratory of Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jiahui An
- Key Laboratory of Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jiansong Zhang
- Key Laboratory of Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Meifen Wang
- Key Laboratory of Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yufeng Li
- Key Laboratory of Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Ping Jiang
- Key Laboratory of Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
38
|
Shan C, Lu Z, Li Z, Sheng H, Fan J, Qi Q, Liu S, Zhang S. 4-hydroxyphenylpyruvate dioxygenase promotes lung cancer growth via pentose phosphate pathway (PPP) flux mediated by LKB1-AMPK/HDAC10/G6PD axis. Cell Death Dis 2019; 10:525. [PMID: 31285420 PMCID: PMC6614486 DOI: 10.1038/s41419-019-1756-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 06/17/2019] [Accepted: 06/18/2019] [Indexed: 02/06/2023]
Abstract
4-hydroxyphenylpyruvate dioxygenase (HPD) is an important modifier of tyrosine metabolism. However, the precise contribution of HPD to cancer metabolism and tumorigenesis remains unclear. In this study, we found that HPD was highly expressed in lung cancer and its higher expression correlated with poor prognosis in lung cancer patients. Suppressed HPD expression was sufficient to decrease oxidative pentose phosphate pathway (PPP) flux, leading to reduced RNA biosynthesis and enhanced reactive oxygen species (ROS) level, attenuated cancer cell proliferation, and tumor growth. Mechanistically, HPD not only promotes tyrosine catabolism leading to increased acetyl-CoA levels, the source of histone acetylation, but also stimulates histone deacetylase 10 (HDAC10) translocation from the nucleus into the cytoplasm mediated by tumor suppressor liver kinase B1 (LKB1)-AMP-activated protein kinase (AMPK) signaling. Both controlled histone acetylation modification, which enhanced transcription of the important PPP enzyme Glucose-6-Phosphate Dehydrogenase (G6PD). Thus, this study reveals HPD as a novel regulator of LKB1-AMPK signaling-mediated HDAC10 nuclear location, which contributes to G6PD expression in promoting tumor growth, which is a promising target for lung cancer treatment.
Collapse
Affiliation(s)
- Changliang Shan
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, 300350, Tianjin, China.
- The First Affiliated Hospital, Biomedical Translational Research Institute, Jinan University, 510632, Guangzhou, Guangdong, China.
| | - Zhaoliang Lu
- Department of Clinical Biological Resource Bank, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, Guangdong, China
| | - Zhen Li
- The First Affiliated Hospital, Biomedical Translational Research Institute, Jinan University, 510632, Guangzhou, Guangdong, China
| | - Hao Sheng
- The First Affiliated Hospital, Biomedical Translational Research Institute, Jinan University, 510632, Guangzhou, Guangdong, China
| | - Jun Fan
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, 510632, Guangzhou, Guangdong, China
| | - Qi Qi
- Department of Pharmacology, School of Medicine, Jinan University, 510632, Guangzhou, Guangdong, China
| | - Shuangping Liu
- Department of Pathology, Medical School, Dalian University, 116622, Dalian, Liaoning, China.
| | - Shuai Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, 301617, Tianjin, China.
| |
Collapse
|
39
|
Fumarate hydratase in cancer: A multifaceted tumour suppressor. Semin Cell Dev Biol 2019; 98:15-25. [PMID: 31085323 DOI: 10.1016/j.semcdb.2019.05.002] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 05/03/2019] [Accepted: 05/06/2019] [Indexed: 02/07/2023]
Abstract
Cancer is now considered a multifactorial disorder with different aetiologies and outcomes. Yet, all cancers share some common molecular features. Among these, the reprogramming of cellular metabolism has emerged as a key player in tumour initiation and progression. The finding that metabolic enzymes such as fumarate hydratase (FH), succinate dehydrogenase (SDH) and isocitrate dehydrogenase (IDH), when mutated, cause cancer suggested that metabolic dysregulation is not only a consequence of oncogenic transformation but that it can act as cancer driver. However, the mechanisms underpinning the link between metabolic dysregulation and cancer remain only partially understood. In this review we discuss the role of FH loss in tumorigenesis, focusing on the role of fumarate as a key activator of a variety of oncogenic cascades. We also discuss how these alterations are integrated and converge towards common biological processes. This review highlights the complexity of the signals elicited by FH loss, describes that fumarate can act as a bona fide oncogenic event, and provides a compelling hypothesis of the stepwise neoplastic progression after FH loss.
Collapse
|
40
|
Kang H, Kim H, Lee S, Youn H, Youn B. Role of Metabolic Reprogramming in Epithelial⁻Mesenchymal Transition (EMT). Int J Mol Sci 2019; 20:ijms20082042. [PMID: 31027222 PMCID: PMC6514888 DOI: 10.3390/ijms20082042] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/08/2019] [Accepted: 04/23/2019] [Indexed: 02/07/2023] Open
Abstract
Activation of epithelial–mesenchymal transition (EMT) is thought to be an essential step for cancer metastasis. Tumor cells undergo EMT in response to a diverse range of extra- and intracellular stimulants. Recently, it was reported that metabolic shifts control EMT progression and induce tumor aggressiveness. In this review, we summarize the involvement of altered glucose, lipid, and amino acid metabolic enzyme expression and the underlying molecular mechanisms in EMT induction in tumor cells. Moreover, we propose that metabolic regulation through gene-specific or pharmacological inhibition may suppress EMT and this treatment strategy may be applied to prevent tumor progression and improve anti-tumor therapeutic efficacy. This review presents evidence for the importance of metabolic changes in tumor progression and emphasizes the need for further studies to better understand tumor metabolism.
Collapse
Affiliation(s)
- Hyunkoo Kang
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea.
| | - Hyunwoo Kim
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea.
| | - Sungmin Lee
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea.
| | - HyeSook Youn
- Department of Integrative Bioscience and Biotechnology, Sejong University, Seoul 05006, Korea.
| | - BuHyun Youn
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea.
- Department of Biological Sciences, Pusan National University, Busan 46241, Korea.
| |
Collapse
|
41
|
Weidle UH, Birzele F, Nopora A. MicroRNAs as Potential Targets for Therapeutic Intervention With Metastasis of Non-small Cell Lung Cancer. Cancer Genomics Proteomics 2019; 16:99-119. [PMID: 30850362 PMCID: PMC6489690 DOI: 10.21873/cgp.20116] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 02/11/2019] [Accepted: 02/12/2019] [Indexed: 02/08/2023] Open
Abstract
The death toll of non-small cell lung cancer (NSCLC) patients is primarily due to metastases, which are poorly amenable to therapeutic intervention. In this review we focus on miRs associated with metastasis of NSCLC as potential new targets for anti-metastatic therapy. We discuss miRs validated as therapeutic targets by in vitro data, identification of target(s) and pathway(s) and in vivo efficacy data in at least one clinically-relevant metastasis-related model. A few of the discussed miRs correlate with the clinical status of NSCLC patients. Using miRs as therapeutic agents has the advantage that targeting a single miR can potentially interfere with several metastatic pathways. Depending on their mode of action, the corresponding miRs can be up- or down-regulated compared to normal matching tissues. Here, we describe therapeutic approaches for reconstitution therapy and miR inhibition, general principles of anti-metastatic therapy as well as current technical pitfalls.
Collapse
Affiliation(s)
- Ulrich H Weidle
- Roche Innovation Center Munich, Roche Diagnostics GmbH, Penzberg, Germany
| | - Fabian Birzele
- Roche Innovation Center Basel, F. Hofman La Roche, Basel, Switzerland
| | - Adam Nopora
- Roche Innovation Center Munich, Roche Diagnostics GmbH, Penzberg, Germany
| |
Collapse
|
42
|
Sheng Z, Xu Y, Li F, Wang S, Huang T, Lu P. CSN5 attenuates Ang II-induced cardiac hypertrophy through stabilizing LKB1. Exp Cell Res 2019; 376:11-17. [PMID: 30710502 DOI: 10.1016/j.yexcr.2019.01.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 01/07/2019] [Accepted: 01/29/2019] [Indexed: 12/18/2022]
Abstract
CSN5 is a critical subunit of the COP9 signalosome (CSN) and has been involved in various cellular processes, but little is known about the role of CSN5 in cardiac disease. In the present study, we found that the expression of CSN5 was increased in Angiotensin II (Ang II)-induced cardiac hypertrophic mice hearts and Ang II-treated cardiomyocytes. We also observed that overexpression of CSN5 significantly inhibited Ang II-induced cardiac hypertrophy, whereas CSN5 silence exhibited the opposite phenotypes. Further investigation demonstrated that CSN5 maintained the activity of AMP-activated protein kinase (AMPK) in cardiomyocyte by enhancement of LKB1. Mechanistically, we found that CSN5 directly interacted and deubiquitinated LKB1 for its stabilization in cardiomyocytes. Finally, our results demonstrated that the anti-hypertrophic effect of CSN5 was partially dependent on stabilization of LKB1. Collectively, these findings suggested that strategies based on activation of CSN5/LKB1 axis might be promising in the treatment of hypertrophic cardiomyopathy.
Collapse
Affiliation(s)
- Zhiyong Sheng
- Intensive Care Unit, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Yun Xu
- Department of emergency, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Fuxin Li
- Intensive Care Unit, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Shu Wang
- Intensive Care Unit, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Tieqiu Huang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Peng Lu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China.
| |
Collapse
|
43
|
He W, Li J. Exendin-4 enhances radiation response of prostate cancer. Prostate 2018; 78:1125-1133. [PMID: 30009503 DOI: 10.1002/pros.23687] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 06/22/2018] [Indexed: 12/27/2022]
Abstract
BACKGROUND Exendin-4, one of the most widely used antidiabetic drugs, has recently been reported to have potential antitumor effects in cancers. Prostate cancer (PC) is one of the most common cancers in male patients with type 2 diabetes mellitus, and radiotherapy plays a vital role in the therapy of PC. Whether exendin-4 has the potential to enhance PC response to ionizing radiation (IR) remains unknown. We aimed to explore whether exendin-4 radiosensitizes PC cells. METHODS GLP-1 receptor (GLP-1R) expression in PC tissue samples and cell lines were analyzed, Human prostate cancer cells (PC3 and LNCap) were treated with IR and exendin-4, and subjected to proliferation, clone formation, cell cycle, immunoblotting, and immunohistochemical analysis. An in situ prostate tumor of animal model was established. RESULTS We found that GLP-1R was expressed in human PC tissues and cell lines. 1-100 nM exendin-4 promoted the anti-proliferation effects of IR in vitro and in vivo, and enhanced radiation-induced G2/M cycle arrest in PC cells in a dose-dependent manner. Furthermore, Ex-4 increased AMPK phosphorylation, decrease the levels of p-mTOR, cyclin B, and p34cdc2 . CONCLUSIONS Our study suggested exendin-4 radiosensitizes PC cells via activation of AMPK A and subsequent inhibition of p-mTOR, cyclin B, and p34cdc2 activation.
Collapse
Affiliation(s)
- Wenjing He
- Institute of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Junhe Li
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
44
|
Ortega-Bernal D, La Rosa CHGD, Arechaga-Ocampo E, Alvarez-Avitia MA, Moreno NS, Rangel-Escareño C. A meta-analysis of transcriptome datasets characterizes malignant transformation from melanocytes and nevi to melanoma. Oncol Lett 2018; 16:1899-1911. [PMID: 30008882 DOI: 10.3892/ol.2018.8861] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 03/12/2018] [Indexed: 12/24/2022] Open
Abstract
Melanoma represents one of the most aggressive malignancies and has a high tendency to metastasize. The present study aims to investigate the molecular mechanisms of two pathways to cancer transformation with the purpose of identifying potential biomarkers. Our approach is based on a meta-analysis of gene expression profiling contrasting two scenarios: A model that describes a transformation pathway from melanocyte to melanoma and a second model where transformation occurs through an intermediary nevus. Data consists of three independent, publicly available microarray datasets from the Gene Expression Omnibus (GEO) database comprising samples from melanocytes, nevi and melanoma. The present analysis identified 808 differentially expressed genes (528 upregulated and 360 downregulated) in melanoma compared with nevi, and 2,331 differentially expressed genes (946 upregulated and 1,385 downregulated) in melanoma compared with melanocytes. Further analysis narrowed down this list, since 682 differentially expressed genes were found in both models (417 upregulated and 265 downregulated). Enrichment analysis identified relevant dysregulated pathways. This article also presented a discussion on significant genes including ADAM like decysin 1, neudesin neurotrophic factor, MMP19, apolipoprotein L6, C-X-C motif chemokine ligand (CXCL)8, basic, immunoglobulin-like variable motif containing and CXCL16. These are of particular interest because they encode secreted proteins hence represent potential blood biomarkers for the early detection of malignant transformation in both scenarios. Cytotoxic T-lymphocyte associated protein 4, an important therapeutic target in melanoma treatment, was also upregulated in both comparisons indicating a potential involvement in immune tolerance, not only at advanced stages but also during the early transformation to melanoma. The results of the present study may provide a research direction for studying the mechanisms underlying the development of melanoma, depending on its origin.
Collapse
Affiliation(s)
- Daniel Ortega-Bernal
- Natural Sciences Department, Universidad Autónoma Metropolitana, Mexico City 05300, Mexico
| | | | - Elena Arechaga-Ocampo
- Natural Sciences Department, Universidad Autónoma Metropolitana, Mexico City 05300, Mexico
| | | | - Nora Sobrevilla Moreno
- Medical Oncology Department, Instituto Nacional de Cancerología, Mexico City 14080, Mexico
| | - Claudia Rangel-Escareño
- Computational and Integrative Genomics Laboratory, Instituto Nacional de Medicina Genómica, Mexico City 14610, Mexico
| |
Collapse
|
45
|
Icard P, Shulman S, Farhat D, Steyaert JM, Alifano M, Lincet H. How the Warburg effect supports aggressiveness and drug resistance of cancer cells? Drug Resist Updat 2018; 38:1-11. [PMID: 29857814 DOI: 10.1016/j.drup.2018.03.001] [Citation(s) in RCA: 389] [Impact Index Per Article: 55.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 03/09/2018] [Accepted: 03/15/2018] [Indexed: 12/11/2022]
Abstract
Cancer cells employ both conventional oxidative metabolism and glycolytic anaerobic metabolism. However, their proliferation is marked by a shift towards increasing glycolytic metabolism even in the presence of O2 (Warburg effect). HIF1, a major hypoxia induced transcription factor, promotes a dissociation between glycolysis and the tricarboxylic acid cycle, a process limiting the efficient production of ATP and citrate which otherwise would arrest glycolysis. The Warburg effect also favors an intracellular alkaline pH which is a driving force in many aspects of cancer cell proliferation (enhancement of glycolysis and cell cycle progression) and of cancer aggressiveness (resistance to various processes including hypoxia, apoptosis, cytotoxic drugs and immune response). This metabolism leads to epigenetic and genetic alterations with the occurrence of multiple new cell phenotypes which enhance cancer cell growth and aggressiveness. In depth understanding of these metabolic changes in cancer cells may lead to the development of novel therapeutic strategies, which when combined with existing cancer treatments, might improve their effectiveness and/or overcome chemoresistance.
Collapse
Affiliation(s)
- Philippe Icard
- Normandie University, UNICAEN, INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment, BioTICLA axis (Biology and Innovative Therapeutics for Ovarian Cancers), Caen, France; UNICANCER, Comprehensive Cancer Center François Baclesse, BioTICLA lab, Caen, France; Department of Thoracic Surgery, University Hospital of Caen, France
| | | | - Diana Farhat
- Inserm U1052, CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon (CRCL), France; Université Lyon Claude Bernard 1, Lyon, France; Department of Chemistry-Biochemistry, Laboratory of Cancer Biology and Molecular Immunology, EDST-PRASE, Lebanese University, Faculty of Sciences, Hadath-Beirut, Lebanon
| | - Jean-Marc Steyaert
- Ecole Polytechnique, Laboratoire d'Informatique (LIX), Palaiseau, France
| | - Marco Alifano
- Department of Thoracic Surgery, Paris Center University Hospital, AP-HP, Paris, France; Paris Descartes University, Paris, France
| | - Hubert Lincet
- Inserm U1052, CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon (CRCL), France; Université Lyon Claude Bernard 1, Lyon, France; ISPB, Faculté de Pharmacie, Lyon, France.
| |
Collapse
|
46
|
Zhang Z, Wang S, Ji D, Qian W, Wang Q, Li J, Gu J, Peng W, Hu T, Ji B, Zhang Y, Wang S, Sun Y. Construction of a ceRNA network reveals potential lncRNA biomarkers in rectal adenocarcinoma. Oncol Rep 2018; 39:2101-2113. [PMID: 29512732 PMCID: PMC5928764 DOI: 10.3892/or.2018.6296] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 02/23/2018] [Indexed: 02/06/2023] Open
Abstract
Competing endogenous RNAs (ceRNAs) render the functions of long non-coding RNAs (lncRNAs) more complicated during cancer processes. Potential lncRNA biomarkers and their roles as ceRNAs have not been clearly described for rectal adenocarcinoma (READ). In the present study, we extracted data from The Cancer Genome Atlas (TCGA) including data from 167 tumor samples and 10 adjacent non-tumor samples. A total of 202 lncRNAs, 190 microRNAs (miRNAs) and 1,530 mRNAs were identified as READ-specific RNAs [log2(fold-change)>2, FDR<0.01]. The Gene Ontology (GO) biological processes and the Kyoto Encyclopaedia of Genes and Genomes (KEGG) pathways were analysed for 1,530 specific mRNAs. Among 202 READ-specific lncRNAs, 7 lncRNAs were identified as being associated with overall survival of READ patients. Then, a ceRNA network was constructed with 34 key lncRNAs, 25 miRNAs and 65 mRNAs. A total of 7 lncRNAs from the network were revealed to be linked to clinical features. The results of qRT-PCR ascertained that our analysis was credible. Overall, this research provides a novel perspective from which to study the lncRNA-related ceRNA network in READ and assists in the identification of new potential biomarkers to be used for diagnostic and prognostic purposes.
Collapse
Affiliation(s)
- Zhiyuan Zhang
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Sen Wang
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Dongjian Ji
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Wenwei Qian
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Qingyuan Wang
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Jie Li
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Jiou Gu
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Wen Peng
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Tao Hu
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Bing Ji
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yue Zhang
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Shijia Wang
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yueming Sun
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
47
|
Cheng J, Shuai X, Gao J, Cai M, Wang G, Tao K. Prognostic significance of AMPK in human malignancies: A meta-analysis. Oncotarget 2018; 7:75739-75748. [PMID: 27716618 PMCID: PMC5342774 DOI: 10.18632/oncotarget.12405] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 09/20/2016] [Indexed: 12/25/2022] Open
Abstract
Background AMPK is a well-investigated kinase mediating cellular metabolism and stress responses. However, its indicative role in survival prognosis remains ill-defined. Therefore we performed this meta-analysis in order to clarify the prognostic impact of AMPK expression in human malignancies. Methods Literatures were retrieved via searching databases of PubMed, Web of Science, Embase and Cochrane Library. Studies comparing the prognostic significance between different AMPK levels among human malignancies were included into the pooled analysis. The statistical procedures were conducted by Review Manager 5.3 and the effect size was displayed by model of odds ratio. Subgroup analyses were additionally implemented to disclose the potential confounding elements. The outcome stability was examined by sensitivity analysis, and both Begg's test and Egger's test were utilized to detect the publication bias across the included studies. Results 21 retrospective cohorts were eventually obtained with a total sample-size of 9987 participants. Patients with higher AMPK expression had better outcomes of 3-year overall survival (P<0.0001), 5-year overall survival (P<0.0001), 10-year overall survival (P<0.0001), 3-year disease free survival (P<0.0001), 5-year disease free survival (P=0.002) and 10-year disease free survival (P=0.0004). Moreover, the majority of subgroup results also verified the favorably prognostic significance of AMPK over-expression. The outcome stability was confirmed by sensitivity analysis. Results of Begg's (P=0.76) and Egger's test (P=0.09) suggested that there was no publication bias within the included trials. Conclusions Higher expression of AMPK significantly indicates better prognosis in human malignancies.
Collapse
Affiliation(s)
- Ji Cheng
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoming Shuai
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinbo Gao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming Cai
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guobin Wang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
48
|
Han YH, Kee JY, Hong SH. Rosmarinic Acid Activates AMPK to Inhibit Metastasis of Colorectal Cancer. Front Pharmacol 2018; 9:68. [PMID: 29459827 PMCID: PMC5807338 DOI: 10.3389/fphar.2018.00068] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 01/18/2018] [Indexed: 12/16/2022] Open
Abstract
Rosmarinic acid (RA) has been used as an anti-inflammatory, anti-diabetic, and anti-cancer agent. Although RA has also been shown to exert an anti-metastatic effect, the mechanism of this effect has not been reported to be associated with AMP-activated protein kinase (AMPK). The aim of this study was to elucidate whether RA could inhibit the metastatic properties of colorectal cancer (CRC) cells via the phosphorylation of AMPK. RA inhibited the proliferation of CRC cells through the induction of cell cycle arrest and apoptosis. In several metastatic phenotypes of CRC cells, RA regulated epithelial-mesenchymal transition (EMT) through the upregulation of an epithelial marker, E-cadherin, and the downregulation of the mesenchymal markers, N-cadherin, snail, twist, vimentin, and slug. Invasion and migration of CRC cells were inhibited and expressions of matrix metalloproteinase (MMP)-2 and MMP-9 were decreased by RA treatment. Adhesion and adhesion molecules such as ICAM-1 and integrin β1 expressions were also reduced by RA treatment. In particular, the effects of RA on EMT and MMPs expressions were due to the activation of AMPK. Moreover, RA inhibited lung metastasis of CRC cells by activating AMPK in mouse model. Collectively, these results proved that RA could be potential therapeutic agent against metastasis of CRC.
Collapse
Affiliation(s)
- Yo-Han Han
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, Iksan, South Korea
| | - Ji-Ye Kee
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, Iksan, South Korea
| | - Seung-Heon Hong
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, Iksan, South Korea
| |
Collapse
|
49
|
Zarredar H, Ansarin K, Baradaran B, Ahdi Khosroshahi S, Farajnia S. Potential Molecular Targets in the Treatment of Lung Cancer Using siRNA Technology. Cancer Invest 2018; 36:37-58. [DOI: 10.1080/07357907.2017.1416393] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Habib Zarredar
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Science, Tabriz, Iran
- Students Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khalil Ansarin
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Science, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Safar Farajnia
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Science, Tabriz, Iran
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
50
|
He SS, Chen Y, Wang HZ, Shen XM, Sun P, Dong J, Liao XB, Guo GF, Chen JG, Xia LP, Hu PL, Qiu HJ, Liu SS, Zhou YX, Wang W, Hu WH, Cai XY. Loss of LKB1 Expression Decreases the Survival and Promotes Laryngeal Cancer Metastasis. J Cancer 2017; 8:3548-3554. [PMID: 29151940 PMCID: PMC5687170 DOI: 10.7150/jca.20535] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 09/07/2017] [Indexed: 12/15/2022] Open
Abstract
Background: Given recent results indicating that diminished LKB1 expression in laryngeal cancer correlates with shorter survival. We aim to perform an analysis estimate the role of decreased liver kinase B1(LKB1) and in the prognostication of human laryngeal squamous cell carcinoma (LSCC). Methods: We conducted a retrospective study and evaluate the expression of LKB1 and p16INK4a (p16) in 208 clinical advanced-stage LSCC tissue samples by using immunohistochemistry. The specimens were received at Sun Yat-sen University Cancer Center (Guangzhou, China). To evaluate the independent prognostic relevance of LKB1, univariate and multivariate Cox regression models were used, overall survival (OS) and distant metastasis-free survival (DMFS) were compared using the Kaplan-Meier method. Results: Immunohistochemical analyses revealed that 80/208 (38.5%) of the LSCC tissue samples expressed high LKB1. Low LKB1 expression was associated with a significantly shorter OS and DMFS than high LKB1 expression (P = 0.041 and 0.028, respectively; log-rank test), and there was a poorer OS in the p16-positive than p16-negative group. In the subgroup stratified by p16 status, the shorter OS were also seen with low LKB1 expression. Multivariate survival analysis indicated that high LKB1 expression was an independent prognostic factor for OS (hazard ratio [HR]: 1.628, 95% confidence interval [CI]: 1.060-2.500, P = 0.026) and DMFS (HR: 2.182, 95% CI: 1.069-4.456, P = 0.032). Conclusions: Our data indicated that low expression of LKB1 was significantly associated with poor prognosis and it may represent a marker of tumor metastasis in patients with LSCC. When combined with p16, LKB1 was also of prognostic value.
Collapse
Affiliation(s)
- Sha-Sha He
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou.,Department of Radiation, Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Yong Chen
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou.,Department of Radiation, Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Hong-Zhi Wang
- Department of Radiation Oncology, Cancer Hospital, Chinese Academy of Medical Science, Peking Union Medical College, Beijing 100021, China
| | - Xiao-Ming Shen
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou.,Department of Radiology, The First People's Hospital of Foshan (The affiliated Foshan Hospital of Sun Yat-Sen University), Foshan, Guangdong, China
| | - Peng Sun
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou.,Department of Pathology, Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Jun Dong
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou.,Department of VIP Region, Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Xin-Biao Liao
- Department of Radiation Oncology, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Gui-Fang Guo
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou.,Department of VIP Region, Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Ju-Gao Chen
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou
| | - Liang-Ping Xia
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou.,Department of VIP Region, Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Pei-Li Hu
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou.,Department of VIP Region, Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Hui-Juan Qiu
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou.,Department of VIP Region, Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Shou-Sheng Liu
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou.,Department of VIP Region, Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Yi-Xin Zhou
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou.,Department of VIP Region, Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Wei Wang
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou.,Department of Gastric Surgery, Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Wei-Han Hu
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou.,Department of Radiation, Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Xiu-Yu Cai
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou.,Department of VIP Region, Cancer Center, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|