1
|
Askari N, Hadizadeh M, Sina M, Parvizpour S, Mousavi SZ, Shamsir MS. Investigating the function and targeting of MET protein as an oncogene kinase in pancreatic ductal adenocarcinoma: A microarray data integration. BIOIMPACTS : BI 2024; 15:30187. [PMID: 40161938 PMCID: PMC11954745 DOI: 10.34172/bi.30187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 05/06/2024] [Accepted: 08/03/2024] [Indexed: 04/02/2025]
Abstract
Introduction Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal disease with a poor prognosis. Kinase proteins are essential regulators of cellular processes and potential targets for drug development. Methods Integration of multiple microarray datasets was screened to find differentially expressed kinases (DE-Kinases) across adjacent normal and tumor tissue samples in PDAC. The most effective kinase for drug design and docking in this study was selected by investigating biological mechanisms and survival analyses. Forty phytochemicals were extracted from the yellow sweet clover, Melilotus officinalis (Linn.) Pall, and were then subjected to in silico screening and molecular docking studies against a specific potent kinase. Results MET, PAK3, and PDK4 were identified as the DE-Kinases. After examining the pathways and biological processes, up-regulated MET had the most significant survival analysis and became our primary kinase for drug design and docking in this study. Four of the extracted phytocompounds of Melilotus officinalis (Linn.) Pall that exhibited high binding affinities with MET and were selected for toxicity analysis. Finally, the stability and mobility of the two nontoxic compounds that passed the toxicity test (dicumarol PubChem CID: 54676038 and melilotigenin PubChem CID: 14059499) were studied by molecular dynamics simulation. Conclusion This study's results identified two phytochemicals in yellow sweet clover that could be used to develop an anticancer drug, but experimental evaluation is necessary to confirm their efficacy.
Collapse
Affiliation(s)
- Nahid Askari
- Department of Biotechnology, Institute of Sciences and High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran
| | - Morteza Hadizadeh
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Sina
- A. Nocivelli Institute for Molecular Medicine, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Sepideh Parvizpour
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyedeh Zahra Mousavi
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohd Shahir Shamsir
- Bioinformatics Research Group (BIRG), Department of Biosciences, Faculty of Science, Universiti Teknologi Malaysia, Skudai, Johor, Malaysia
| |
Collapse
|
2
|
Si T, Huang L, Liang T, Huang P, Zhang H, Zhang M, Zhou X. Ruangan Lidan decoction inhibits the growth and metastasis of liver cancer by downregulating miR-9-5p and upregulating PDK4. Cancer Biol Ther 2023; 24:2246198. [PMID: 37773732 PMCID: PMC10543352 DOI: 10.1080/15384047.2023.2246198] [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: 02/06/2023] [Accepted: 08/04/2023] [Indexed: 10/01/2023] Open
Abstract
A growing number of studies have suggested that traditional Chinese medicine (TCM) plays an essential role in the development and occurrence of liver cancer. However, the function of Ruangan Lidan decoction (RLD) in liver cancer are not yet adequately identified and manifested, which attracted our attention. The key genes related to liver cancer and RLD and the upstream miRNAs of PDK4 were obtained based on bioinformatics analysis, followed by verification of the targeting relationship between miR-9-5p and PDK4. Next, Huh7 cells were treated with RLD to detect cell proliferation, colony formation, migration, invasion, and apoptosis by multiple assays with gain- and loss-of-function experiments. Moreover, subcutaneous transplanted tumor model and lung metastasis model of liver cancer in nude mice were established to further verify the functional role of RLD in liver cancer growth and metastasis via miR-9-5p/PDK4 axis. Bioinformatics analysis found that PDK4 and miR-9-5p were related to liver cancer, and PDK4 may be a downstream regulator of RLD. miR-9-5p could target and inhibit PDK4. In vitro cell experiments demonstrated that RLD suppressed liver cancer cell proliferation, invasion and migration, and promoted apoptosis by inhibiting miR-9-5p expression and promoting PDK4 expression. In vivo animal experiments further confirmed that RLD inhibited liver cancer growth and metastasis via upregulation of miR-9-5p-dependent PDK4. RLD downregulated miR-9-5p and upregulated PDK4 to inhibit the proliferation, migration, invasion, and induce apoptosis, thereby suppressing the growth and metastasis of liver cancer, highlighting a potential novel target for treatment of liver cancer.
Collapse
Affiliation(s)
- Tao Si
- Department of Oncology, Liuzhou Traditional Chinese Medical Hospital, Liuzhou, China
| | - Liyin Huang
- Graduate school, Guangxi University of Chinese Medicine, Nanning, China
| | - Ting Liang
- Graduate school, Guangxi University of Chinese Medicine, Nanning, China
| | - Ping Huang
- Department of Oncology, Liuzhou Traditional Chinese Medical Hospital, Liuzhou, China
| | - Hongyu Zhang
- Department of Clinical Laboratory, Liuzhou Traditional Chinese Medical Hospital, Liuzhou, China
| | - Mingmin Zhang
- Department of Oncology, Liuzhou Traditional Chinese Medical Hospital, Liuzhou, China
| | - Xiaoling Zhou
- Department of Gastroenterology, Liuzhou Traditional Chinese Medical Hospital, Liuzhou, China
| |
Collapse
|
3
|
Huang Y, Liangpunsakul S, Rudraiah S, Ma J, Keshipeddy SK, Wright D, Costa A, Burgess D, Zhang Y, Huda N, Wang L, Yang Z. HMGB2 is a potential diagnostic marker and therapeutic target for liver fibrosis and cirrhosis. Hepatol Commun 2023; 7:e0299. [PMID: 37930124 PMCID: PMC10629741 DOI: 10.1097/hc9.0000000000000299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 08/23/2023] [Indexed: 11/07/2023] Open
Abstract
BACKGROUND High mobility group proteins 1 and 2 (HMGB1 and HMGB2) are 80% conserved in amino acid sequence. The function of HMGB1 in inflammation and fibrosis has been extensively characterized. However, an unaddressed central question is the role of HMGB2 on liver fibrosis. In this study, we provided convincing evidence that the HMGB2 expression was significantly upregulated in human liver fibrosis and cirrhosis, as well as in several mouse liver fibrosis models. METHODS The carbon tetrachloride (CCl4) induced liver fibrosis mouse model was used. AAV8-Hmgb2 was utilized to overexpress Hmgb2 in the liver, while Hmgb2-/- mice were used for loss of function experiments. The HMGB2 inhibitor inflachromene and liposome-shHMGB2 (lipo-shHMGB2) were employed for therapeutic intervention. RESULTS The serum HMGB2 levels were also markedly elevated in patients with liver fibrosis and cirrhosis. Deletion of Hmgb2 in Hmgb2-/- mice or inhibition of HMGB2 in mice using a small molecule ICM slowed the progression of CCl4-induced liver fibrosis despite constant HMGB1 expression. In contrast, AAV8-mediated overexpression of Hmgb2 enchanced CCl4-incuded liver fibrosis. Primary hepatic stellate cells (HSCs) isolated from Hmgb2-/- mice showed significantly impaired transdifferentiation and diminished activation of α-SMA, despite a modest induction of HMGB1 protein. RNA-seq analysis revealed the induction of top 45 CCl4-activated genes in multiple signaling pathways including integrin signaling and inflammation. The activation of these genes by CCl4 were abolished in Hmgb2-/- mice or in ICM-treated mice. These included C-X3-C motif chemokine receptor 1 (Cx3cr1) associated with inflammation, cyclin B (Ccnb) associated with cell cycle, DNA topoisomerase 2-alpha (Top2a) associated with intracellular component, and fibrillin (Fbn) and fibromodulin (Fmod) associated with extracellular matrix. CONCLUSION We conclude that HMGB2 is indispensable for stellate cell activation. Therefore, HMGB2 may serve as a potential therapeutic target to prevent HSC activation during chronic liver injury. The blood HMGB2 level may also serve as a potential diagnostic marker to detect early stage of liver fibrosis and cirrhosis in humans.
Collapse
Affiliation(s)
- Yi Huang
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut, USA
| | - Suthat Liangpunsakul
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University, Indianapolis, Indiana, USA
- Medicine Service, Roudebush Veterans Administration Medical Center, Indianapolis, Indiana, USA
| | - Swetha Rudraiah
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut, USA
| | - Jing Ma
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University, Indianapolis, Indiana, USA
| | - Santosh K. Keshipeddy
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut, USA
| | - Dennis Wright
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut, USA
| | - Antonio Costa
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut, USA
| | - Diane Burgess
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut, USA
| | - Yuxia Zhang
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Nazmul Huda
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University, Indianapolis, Indiana, USA
| | - Li Wang
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona, USA
| | - Zhihong Yang
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University, Indianapolis, Indiana, USA
| |
Collapse
|
4
|
Woolbright BL, Rajendran G, Abbott E, Martin A, Didde R, Dennis K, Harris RA, Taylor JA. Pyruvate Dehydrogenase Kinase 4 Deficiency Increases Tumorigenesis in a Murine Model of Bladder Cancer. Cancers (Basel) 2023; 15:1654. [PMID: 36980540 PMCID: PMC10046149 DOI: 10.3390/cancers15061654] [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: 01/26/2023] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/10/2023] Open
Abstract
Pyruvate dehydrogenase kinase 4 (PDK4) is a mitochondrial isozyme in the PDK family (PDK1-4) partially responsible for phosphorylation of pyruvate dehydrogenase (PDH). Phosphorylation of PDH is thought to result in a pro-proliferative shift in metabolism that sustains growth of cancer cells. Previous data from our lab indicate the pan-PDK inhibitor dichloroacetate (DCA) or acute genetic knockdown of PDK4 blocks proliferation of bladder cancer (BCa) cells. The goal of this study was to determine the role of PDK4 in an in vivo BCa model, with the hypothesis that genetic depletion of PDK4 would impair formation of BCa. PDK4-/- or WT animals were exposed to N-Butyl-N-(4-hydroxybutyl) nitrosamine (BBN) for 16 weeks, and tumors were allowed to develop for up to 7 additional weeks. PDK4-/- mice had significantly larger tumors at later time points. When animals were treated with cisplatin, PDK4-/- animals still had larger tumors than WT mice. PDK4 expression was assessed in human tissue and in mice. WT mice lost expression of PDK4 as tumors became muscle-invasive. Similar results were observed in human samples, wherein tumors had less expression of PDK4 than benign tissue. In summary, PDK4 has a complex, multifunctional role in BCa and may represent an underrecognized tumor suppressor.
Collapse
Affiliation(s)
| | - Ganeshkumar Rajendran
- Department of Urology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Erika Abbott
- Department of Urology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Austin Martin
- School of Medicine, Kansas University Medical Center, Kansas City, KS 66160, USA
| | - Ryan Didde
- School of Medicine, Kansas University Medical Center, Kansas City, KS 66160, USA
| | - Katie Dennis
- Department of Pathology, Kansas University Medical Center, Kansas City, KS 66160, USA
| | - Robert A. Harris
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - John A. Taylor
- Department of Urology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
5
|
Stacpoole PW, McCall CE. The pyruvate dehydrogenase complex: Life's essential, vulnerable and druggable energy homeostat. Mitochondrion 2023; 70:59-102. [PMID: 36863425 DOI: 10.1016/j.mito.2023.02.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 01/30/2023] [Accepted: 02/13/2023] [Indexed: 03/04/2023]
Abstract
Found in all organisms, pyruvate dehydrogenase complexes (PDC) are the keystones of prokaryotic and eukaryotic energy metabolism. In eukaryotic organisms these multi-component megacomplexes provide a crucial mechanistic link between cytoplasmic glycolysis and the mitochondrial tricarboxylic acid (TCA) cycle. As a consequence, PDCs also influence the metabolism of branched chain amino acids, lipids and, ultimately, oxidative phosphorylation (OXPHOS). PDC activity is an essential determinant of the metabolic and bioenergetic flexibility of metazoan organisms in adapting to changes in development, nutrient availability and various stresses that challenge maintenance of homeostasis. This canonical role of the PDC has been extensively probed over the past decades by multidisciplinary investigations into its causal association with diverse physiological and pathological conditions, the latter making the PDC an increasingly viable therapeutic target. Here we review the biology of the remarkable PDC and its emerging importance in the pathobiology and treatment of diverse congenital and acquired disorders of metabolic integration.
Collapse
Affiliation(s)
- Peter W Stacpoole
- Department of Medicine (Division of Endocrinology, Metabolism and Diabetes), and Department of Biochemistry and Molecular Biology, University of Florida, College of Medicine, Gainesville, FL, United States.
| | - Charles E McCall
- Department of Internal Medicine and Translational Sciences, and Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
6
|
Bu Y, Hao J, He J, Li X, Liu Y, Ma L. Tumor-promoting properties of enolase-phosphatase 1 in breast cancer via activating the NF-κB signaling pathway. Mol Biol Rep 2023; 50:993-1004. [PMID: 36378417 DOI: 10.1007/s11033-022-08066-w] [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: 08/23/2022] [Accepted: 10/31/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Evidence suggests that enolase-phosphatase 1 (ENOPH1) is involved in the progression of some certain types of cancers and acts as an oncogenic factor in tumor progression. The present study aimed to identify the central role of ENOPH1 in the progression of breast cancer (BC), a highly proliferative and aggressive disease. METHODS AND RESULTS ENOPH1 expression in BC tissues was explored based on the online resource and 40 paired fresh BC and para-carcinoma samples. Functional assays were performed to evaluate the biological effect of ENOPH1 on cell proliferation and migration in ENOPH1-silenced or overexpressing BC cell lines. Blockade of NF-κB by BAY11-7082 was performed to evaluate whether ENOPH1 exerted tumor-promoting properties via regulating the NF-κB signaling pathway. Results of the present study demonstrated that ENOPH1 expression was profoundly upregulated in BC tissues compared with adjacent breast tissues, and ENOPH1 expression was associated with cancer stage, node metastasis status, and overall survival. Functional assays demonstrated that ENOPH1 overexpression significantly accelerated BC cell proliferation, migration, and invasion, while genetic knockdown of ENOPH1 yielded the opposite effects. Mechanistically, ENOPH1 activated the NF-κB pathway, as evidenced by increased expression of NF-κB downstream genes and enhanced NF-κB p65 nuclear translocation. Furthermore, the oncogenic properties of ENOPH1 in proliferation, migration, and invasion were restrained following inhibition of the NF-κB signaling pathway. CONCLUSIONS These findings indicated the significance of ENOPH1 in promoting cell proliferation and invasion, mainly through activating the NF-κB pathway, suggesting that ENOPH1 might be an attractive prognostic factor and a potential target for BC therapy.
Collapse
Affiliation(s)
- Yuhui Bu
- Breast Center, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, Hebei, China.,Breast Center, Cangzhou People's Hospital, Cangzhou, Hebei, China
| | - Jun Hao
- Department of Pathology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jianchao He
- Department of Breast Surgery, Affiliated Hospital of Hebei Engineering University, Handan, Hebei, China
| | - Xiaolong Li
- Department of Breast Surgery, The Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
| | - Yinfeng Liu
- Department of Breast Surgery, First Hospital of Qinhuangdao, Qinhuangdao, Hebei, China
| | - Li Ma
- Breast Center, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, Hebei, China.
| |
Collapse
|
7
|
Li W, Long X, Li F, Cao Y, Liu J, Fu S, Guo W, Hu G. Lysine stimulates the development of the murine mammary gland at puberty via PI3K/AKT/mTOR signalling axis. J Anim Physiol Anim Nutr (Berl) 2022; 106:1420-1430. [PMID: 35923149 DOI: 10.1111/jpn.13756] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/21/2022] [Accepted: 07/08/2022] [Indexed: 11/29/2022]
Abstract
Lysine is one of the essential amino acids. The effect of lysine on milk protein and milk fat anabolism has been reported, but the effect on mammary glands development has not been studied in detail. The normal development of the mammary glands at puberty is crucial to lactation of mammals. In this study, to explore the effect of lysine on mammary glands development, we fed different concentrations of lysine (0.025%, 0.05%, 0.1%) to pubertal mice and found that the addition of 0.1% lysine to drinking water significantly promoted mammary glands development. Furthermore, we treated mMECs (mouse mammary epithelial cells) with different concentrations of lysine (0, 0.2, 0.4, 0.6, 0.8 and 1 mM) to explore the underlying mechanism, and found that lysine promoted the proliferation of mMECs and development of mammary glands through PI3K/AKT/mTOR signalling pathway in pubertal mice. Overall, the results of this study revealed that lysine activated the PI3K/AKT/mTOR signal axis, elevated protein concentrations of cell proliferation markers, such as PCNA, Cyclin D1 and D3, and enhanced the proliferation of mMECs, finally promoted the murine mammary glands development at puberty.
Collapse
Affiliation(s)
- Wen Li
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Xiaoyu Long
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Feng Li
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Yu Cao
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Juxiong Liu
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Shoupeng Fu
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Wenjin Guo
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Guiqiu Hu
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| |
Collapse
|
8
|
Zhou Y, Guo Y, Tam KY. Targeting glucose metabolism to develop anticancer treatments and therapeutic patents. Expert Opin Ther Pat 2022; 32:441-453. [PMID: 35001793 DOI: 10.1080/13543776.2022.2027912] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION One of the most distinctive hallmarks of cancer cells is increased glucose consumption for aerobic glycolysis which is named the Warburg effect. In recent decades, extensive research has been carried out to exploit this famous phenomenon, trying to detect promising targetable vulnerabilities in altered metabolism to fight cancer. Targeting aberrant glucose metabolism can perturb cancer malignant proliferation and even induce programmed cell death. AREAS COVERED This review covered the recent patents which focused on targeting key glycolytic enzymes including hexokinase, pyruvate dehydrogenase kinases and lactate dehydrogenase for cancer treatment. EXPERT OPINION Compared with the conventional cancer treatment, specifically targeting the well-known Achilles heel Warburg effect has attracted considerable attention. Although there is still no single glycolytic agent for clinical cancer treatment, the combination of glycolytic inhibitor with conventional anticancer drug or the combined use of multiple glycolytic inhibitors are being investigated extensively in recent years, which could emerge as attractive anticancer strategies.
Collapse
Affiliation(s)
- Yan Zhou
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR, PR China
| | - Yizhen Guo
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR, PR China
| | - Kin Yip Tam
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR, PR China
| |
Collapse
|
9
|
Jane EP, Premkumar DR, Rajasundaram D, Thambireddy S, Reslink MC, Agnihotri S, Pollack IF. Reversing tozasertib resistance in glioma through inhibition of pyruvate dehydrogenase kinases. Mol Oncol 2022; 16:219-249. [PMID: 34058053 PMCID: PMC8732347 DOI: 10.1002/1878-0261.13025] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 04/23/2021] [Accepted: 05/28/2021] [Indexed: 12/19/2022] Open
Abstract
Acquired resistance to conventional chemotherapeutic agents limits their effectiveness and can cause cancer treatment to fail. Because enzymes in the aurora kinase family are vital regulators of several mitotic events, we reasoned that targeting these kinases with tozasertib, a pan-aurora kinase inhibitor, would not only cause cytokinesis defects, but also induce cell death in high-grade pediatric and adult glioma cell lines. We found that tozasertib induced cell cycle arrest, increased mitochondrial permeability and reactive oxygen species generation, inhibited cell growth and migration, and promoted cellular senescence and pro-apoptotic activity. However, sustained exposure to tozasertib at clinically relevant concentrations conferred resistance, which led us to examine the mechanistic basis for the emergence of drug resistance. RNA-sequence analysis revealed a significant upregulation of the gene encoding pyruvate dehydrogenase kinase isoenzyme 4 (PDK4), a pyruvate dehydrogenase (PDH) inhibitory kinase that plays a crucial role in the control of metabolic flexibility under various physiological conditions. Upregulation of PDK1, PDK2, PDK3, or PDK4 protein levels was positively correlated with tozasertib-induced resistance through inhibition of PDH activity. Tozasertib-resistant cells exhibited increased mitochondrial mass as measured by 10-N-nonyl-Acridine Orange. Inhibition of PDK with dichloroacetate resulted in increased mitochondrial permeability and cell death in tozasertib-resistant glioma cell lines. Based on these results, we believe that PDK is a selective target for the tozasertib resistance phenotype and should be considered for further preclinical evaluations.
Collapse
Affiliation(s)
- Esther P Jane
- Department of Neurosurgery, University of Pittsburgh School of Medicine, PA, USA
| | - Daniel R Premkumar
- Department of Neurosurgery, University of Pittsburgh School of Medicine, PA, USA
- Department of Neurosurgery, UPMC Hillman Cancer Center, PA, USA
| | | | - Swetha Thambireddy
- Department of Neurosurgery, University of Pittsburgh School of Medicine, PA, USA
| | - Matthew C Reslink
- Department of Neurosurgery, University of Pittsburgh School of Medicine, PA, USA
| | - Sameer Agnihotri
- Department of Neurosurgery, University of Pittsburgh School of Medicine, PA, USA
- Department of Neurosurgery, UPMC Hillman Cancer Center, PA, USA
| | - Ian F Pollack
- Department of Neurosurgery, University of Pittsburgh School of Medicine, PA, USA
- Department of Neurosurgery, UPMC Hillman Cancer Center, PA, USA
| |
Collapse
|
10
|
Oesch F, Fruth D, Hengstler JG, Fabian E, Berger FI, Landsiedel R. Enigmatic mechanism of the N-vinylpyrrolidone hepatocarcinogenicity in the rat. Arch Toxicol 2021; 95:3717-3744. [PMID: 34595563 PMCID: PMC8536644 DOI: 10.1007/s00204-021-03151-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 08/26/2021] [Indexed: 11/24/2022]
Abstract
N-vinyl pyrrolidone (NVP) is produced up to several thousand tons per year as starting material for the production of polymers to be used in pharmaceutics, cosmetics and food technology. Upon inhalation NVP was carcinogenic in the rat, liver tumor formation is starting already at the rather low concentration of 5 ppm. Hence, differentiation whether NVP is a genotoxic carcinogen (presumed to generally have no dose threshold for the carcinogenic activity) or a non-genotoxic carcinogen (with a potentially definable threshold) is highly important. In the present study, therefore, the existing genotoxicity investigations on NVP (all showing consistently negative results) were extended and complemented with investigations on possible alternative mechanisms, which also all proved negative. All tests were performed in the same species (rat) using the same route of exposure (inhalation) and the same doses of NVP (5, 10 and 20 ppm) as had been used in the positive carcinogenicity test. Specifically, the tests included an ex vivo Comet assay (so far not available) and an ex vivo micronucleus test (in contrast to the already available micronucleus test in mice here in the same species and by the same route of application as in the bioassay which had shown the carcinogenicity), tests on oxidative stress (non-protein-bound sulfhydryls and glutathione recycling test), mechanisms mediated by hepatic receptors, the activation of which had been shown earlier to lead to carcinogenicity in some instances (Ah receptor, CAR, PXR, PPARα). No indications were obtained for any of the investigated mechanisms to be responsible for or to contribute to the observed carcinogenicity of NVP. The most important of these exclusions is genotoxicity. Thus, NVP can rightfully be regarded and treated as a non-genotoxic carcinogen and threshold approaches to the assessment of this chemical are supported. However, the mechanism underlying the carcinogenicity of NVP in rats remains unclear.
Collapse
Affiliation(s)
- Franz Oesch
- Oesch-Tox Toxicological Consulting and Expert Opinions GmbH&CoKG, Rheinblick 21, 55263, Ingelheim, Germany.,Institute of Toxicology, Johannes Gutenberg University, 55131, Mainz, Germany
| | - Daniela Fruth
- Experimental Toxicology and Ecology, BASF SE, 67056, Ludwigshafen am Rhein, Germany.,Knoell Germany GmbH, Eastsite XII, Konrad-Zuse-Ring 25, 68163, Mannheim, Germany
| | - Jan G Hengstler
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), University of Dortmund, Dortmund, Germany
| | - Eric Fabian
- Experimental Toxicology and Ecology, BASF SE, 67056, Ludwigshafen am Rhein, Germany
| | - Franz Ingo Berger
- Regulatory Toxicology Chemicals, BASF SE, 67056, Ludwigshafen am Rhein, Germany
| | - Robert Landsiedel
- Experimental Toxicology and Ecology, BASF SE, 67056, Ludwigshafen am Rhein, Germany.
| |
Collapse
|
11
|
Liu Y, Li YQ, Huang SH, Li YL, Xia JW, Jia JS, Wei F, Wang JH, Dai GQ, Wang YC, Li XY, Han LX, Zhang XL, Xiang XD, Zhao WT, Xiao D, Lin XL. Liver-specific over-expression of Cripto-1 in transgenic mice promotes hepatocyte proliferation and deregulated expression of hepatocarcinogenesis-related genes and signaling pathways. Aging (Albany NY) 2021; 13:21155-21190. [PMID: 34517344 PMCID: PMC8457585 DOI: 10.18632/aging.203402] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 07/13/2021] [Indexed: 11/25/2022]
Abstract
In this study, we investigated the role of embryonic gene Cripto-1 (CR-1) in hepatocellular carcinoma (HCC) using hepatocyte-specific CR-1-overexpressing transgenic mice. The expression of truncated 1.7-kb CR-1 transcript (SF-CR-1) was significantly higher than the full-length 2.0-kb CR-1 transcript (FL-CR-1) in a majority of HCC tissues and cell lines. Moreover, CR-1 mRNA and protein levels were significantly higher in HCC tissues than adjacent normal liver tissues. Hepatocyte-specific over-expression of CR-1 in transgenic mice enhanced hepatocyte proliferation after 2/3 partial hepatectomy (2/3 PHx). CR-1 over-expression significantly increased in vivo xenograft tumor growth of HCC cells in nude mice and in vitro HCC cell proliferation, migration, and invasion. CR-1 over-expression in the transgenic mouse livers deregulated HCC-related signaling pathways such as AKT, Wnt/β-catenin, Stat3, MAPK/ERK, JNK, TGF-β and Notch, as well as expression of HCC-related genes such as CD5L, S100A8, S100A9, Timd4, Orm2, Orm3, PDK4, DMBT1, G0S2, Plk2, Plk3, Gsta1 and Gsta2. However, histological signs of precancerous lesions, hepatocyte dysplasia or HCC formation were not observed in the livers of 3-, 6- or 8-month-old hepatocyte-specific CR-1-overexpressing transgenic mice. These findings demonstrate that liver-specific CR-1 overexpression in transgenic mice deregulates signaling pathways and genes associated with HCC.
Collapse
Affiliation(s)
- Yu Liu
- Cancer Research Institute, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
- Institute of Comparative Medicine and Laboratory Animal Center, Southern Medical University, Guangzhou 510515, China
| | - Yan-Qing Li
- Department of Hematology, Central Hospital of Xuhui District, Shanghai 200030, China
| | - Shi-Hao Huang
- Cancer Research Institute, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| | - Yong-Long Li
- Cancer Research Institute, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
- Institute of Comparative Medicine and Laboratory Animal Center, Southern Medical University, Guangzhou 510515, China
| | - Jia-Wei Xia
- The Third People’s Hospital of Kunming (The Sixth Affiliated Hospital of Dali University), Kunming 650041, China
| | - Jun-Shuang Jia
- Cancer Research Institute, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| | - Fang Wei
- Cancer Research Institute, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| | - Jia-Hong Wang
- Cancer Research Institute, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| | - Guan-Qi Dai
- Cancer Research Institute, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| | - Yu-Cai Wang
- Cancer Research Institute, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| | - Xiao-Yan Li
- Cancer Research Institute, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
- Institute of Comparative Medicine and Laboratory Animal Center, Southern Medical University, Guangzhou 510515, China
| | - Liu-Xin Han
- The Third People’s Hospital of Kunming (The Sixth Affiliated Hospital of Dali University), Kunming 650041, China
| | - Xiao-Ling Zhang
- Department of Physiology, Faculty of Basic Medical Sciences, Guilin Medical University, Guilin 541004, China
| | - Xu-Dong Xiang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming 650118, China
| | - Wen-Tao Zhao
- Department of Gastrointestinal Oncology, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming 650118, China
| | - Dong Xiao
- Cancer Research Institute, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
- Institute of Comparative Medicine and Laboratory Animal Center, Southern Medical University, Guangzhou 510515, China
| | - Xiao-Lin Lin
- Cancer Research Institute, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
12
|
Liu B, Zhang Y, Suo J. Increased Expression of PDK4 Was Displayed in Gastric Cancer and Exhibited an Association With Glucose Metabolism. Front Genet 2021; 12:689585. [PMID: 34220962 PMCID: PMC8248380 DOI: 10.3389/fgene.2021.689585] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 05/12/2021] [Indexed: 01/15/2023] Open
Abstract
Previous studies reported that pyruvate dehydrogenase kinase 4 (PDK4) is closely related to diabetes, heart disease, and carcinomas. Nevertheless, the role of PDK4 in gastric cancer (GC) occurrence and development is yet poorly understood. Our experiments were taken to evaluate PDK4's function in GC. The Cancer Genome Atlas tumor genome map database was employed to validate the levels of PDK family in different grades and stages of GC. The survival ratio of PDK families in GC was detected by the Kaplan-Meier plotter database. The links existing in the expression of PDK family and the level of tumor-infiltrating immune cells were investigated by tumor immunity assessment resource (TIMER). PDK4-associated signal pathways in GC were analyzed by the Kyoto Encyclopedia of Genes and Genomes pathway analysis. PDK4 mRNA level in the GC cells was measured by qRT-PCR. Cell counting kit-8 and Transwell assays were separately carried out to evaluate PDK4-induced influence on GC cell proliferation, migration, and invasion. Our data suggested that GC cells highly expressed PDK4, and PDK4 expression presented a significant relation with the staging, grade, and survival rate of GC. PDK4 expression presented a positive correlation with the types of different infiltrating immune cells, comprising B cells, CD4+ T cells, and dendritic cells. Meanwhile, PDK4 expression exhibited a strong association with macrophages. Survival analysis revealed that the expression of PDK4 displayed a relationship with the prognosis of patients. Therefore, PDK4 was liable to be a biomarker for prognosis. Our results further displayed that PDK4 might modulate the glycolysis level in GC cells, and its expression was associated with GC cell proliferation, migration, and invasion. These data may provide insights into designing a new treatment strategy for GC.
Collapse
Affiliation(s)
| | | | - Jian Suo
- Department of Gastrocolorectal Surgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
13
|
PDK2: An Underappreciated Regulator of Liver Metabolism. LIVERS 2021. [DOI: 10.3390/livers1020008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Pyruvate metabolism is critical for all mammalian cells. The pyruvate dehydrogenase complex couples the pyruvate formed as the primary product of glycolysis to the formation of acetyl-CoA required as the primary substrate of the citric acid cycle. Dysregulation of this coupling contributes to alterations in metabolic flexibility in obesity, diabetes, cancer, and more. The pyruvate dehydrogenase kinase family of isozymes phosphorylate and inactive the pyruvate dehydrogenase complex in the mitochondria. This function makes them critical mediators of mitochondrial metabolism and drug targets in a number of disease states. The liver expresses multiple PDKs, predominantly PDK1 and PDK2 in the fed state and PDK1, PDK2, and PDK4 in the starved and diabetic states. PDK4 undergoes substantial transcriptional regulation in response to a diverse array of stimuli in most tissues. PDK2 has received less attention than PDK4 potentially due to the dramatic changes in transcriptional gene regulation. However, PDK2 is more responsive than the other PDKs to feedforward and feedback regulation by substrates and products of the pyruvate dehydrogenase complex. Although underappreciated, this makes PDK2 particularly important for the minute-to-minute fine control of the pyruvate dehydrogenase complex and a major contributor to metabolic flexibility. The purpose of this review is to characterize the underappreciated role of PDK2 in liver metabolism. We will focus on known biological actions and physiological roles as well as what roles PDK2 may play in disease states. We will also define current inhibitors and address their potential as therapeutic agents in the future.
Collapse
|
14
|
Atas E, Oberhuber M, Kenner L. The Implications of PDK1-4 on Tumor Energy Metabolism, Aggressiveness and Therapy Resistance. Front Oncol 2020; 10:583217. [PMID: 33384955 PMCID: PMC7771695 DOI: 10.3389/fonc.2020.583217] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 11/13/2020] [Indexed: 12/17/2022] Open
Abstract
A metabolic shift from oxidative phosphorylation (OXPHOS) to glycolysis-known as the Warburg effect-is characteristic for many cancers. It gives the cancer cells a survival advantage in the hypoxic tumor microenvironment and protects them from cytotoxic effects of oxidative damage and apoptosis. The main regulators of this metabolic shift are the pyruvate dehydrogenase complex and pyruvate dehydrogenase kinase (PDK) isoforms 1-4. PDK is known to be overexpressed in several cancers and is associated with bad prognosis and therapy resistance. Whereas the expression of PDK1-3 is tissue specific, PDK4 expression is dependent on the energetic state of the whole organism. In contrast to other PDK isoforms, not only oncogenic, but also tumor suppressive functions of PDK4 have been reported. In tumors that profit from high OXPHOS and high de novo fatty acid synthesis, PDK4 can have a protective effect. This is the case for prostate cancer, the most common cancer in men, and makes PDK4 an interesting therapeutic target. While most work is focused on PDK in tumors characterized by high glycolytic activity, little research is devoted to those cases where PDK4 acts protective and is therefore highly needed.
Collapse
Affiliation(s)
- Emine Atas
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Monika Oberhuber
- Department of Pathology, Medical University of Vienna, Vienna, Austria
- Area ‘Data & Technologies’, CBmed—Center for Biomarker Research in Medicine GmbH, Graz, Austria
| | - Lukas Kenner
- Department of Pathology, Medical University of Vienna, Vienna, Austria
- Area ‘Data & Technologies’, CBmed—Center for Biomarker Research in Medicine GmbH, Graz, Austria
- Unit of Pathology of Laboratory Animals, University of Veterinary Medicine Vienna, Vienna, Austria
- Christian Doppler Laboratory for Applied Metabolomics (CDL AM), Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
15
|
Si T, Ning X, Zhao H, Zhang M, Huang P, Hu Z, Yang L, Lin L. microRNA-9-5p regulates the mitochondrial function of hepatocellular carcinoma cells through suppressing PDK4. Cancer Gene Ther 2020; 28:706-718. [PMID: 33257740 DOI: 10.1038/s41417-020-00253-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 10/09/2020] [Accepted: 10/29/2020] [Indexed: 02/06/2023]
Abstract
Due to the lack of early diagnostic and effective treatment modalities, hepatocellular carcinoma (HCC) is still the most lethal cancer with a high mortality on a global scale. Recent studies have highlighted the key roles of microRNAs (miRs) in HCC development. In the study, we attempted to investigate the potential role of miR-9-5p in the progression of HCC. Expression of pyruvate dehydrogenase kinase 4 (PDK4) and miR-9-5p was examined in HCC tissues collected from HCC patients and cell lines. The proliferation, migration, invasion, and apoptosis of HCC cells, and levels of oxygen consumption rate, extracellular acidification rate and reactive oxygen species (ROS) as well as the tumorigenicity of transfected cells in vivo were measured after gain- and loss-of-function experiments in HCC cells. It was revealed that miR-9-5p was upregulated, while PDK4 was poorly expressed in HCC tissues and cells, associating with a poor prognosis of HCC patients. miR-9-5p directly targeted PDK4 and could downregulate its expression, thus leading to promoted cell proliferation, invasion and migration, enhanced mitochondrial activity and energy metabolism, and suppressed apoptosis in HCC cells, along with increased tumorigenicity in mouse xenograft models. Altogether, miR-9-5p facilitated mitochondrial energy metabolism of HCC cells by downregulating PDK4, promoting the development of HCC. miR-9-5p and PDK4 may serve as potential therapeutic targets for preventing recurrence and metastasis of HCC.
Collapse
Affiliation(s)
- Tao Si
- Integrative Cancer Centre, Guangzhou University of Chinese Medicine First Affiliated Hospital, 510405, Guangzhou, China.,Department of Oncology, Liuzhou Traditional Chinese Medical Hospital, 545001, Liuzhou, China
| | - Xuejian Ning
- Department of Oncology, Liuzhou Traditional Chinese Medical Hospital, 545001, Liuzhou, China
| | - Hongwei Zhao
- Integrative Cancer Centre, Guangzhou University of Chinese Medicine First Affiliated Hospital, 510405, Guangzhou, China
| | - Mingmin Zhang
- Guangxi University of Chinese Medicine, 530000, Nanning, China
| | - Ping Huang
- Department of Oncology, Liuzhou Traditional Chinese Medical Hospital, 545001, Liuzhou, China
| | - Zhengguo Hu
- Department of Oncology, Liuzhou Traditional Chinese Medical Hospital, 545001, Liuzhou, China
| | - Liu Yang
- Department of Oncology, Liuzhou Traditional Chinese Medical Hospital, 545001, Liuzhou, China
| | - Lizhu Lin
- Integrative Cancer Centre, Guangzhou University of Chinese Medicine First Affiliated Hospital, 510405, Guangzhou, China.
| |
Collapse
|
16
|
Xu H, He Y, Ma J, Zhao Y, Liu Y, Sun L, Su J. Inhibition of pyruvate dehydrogenase kinase‑1 by dicoumarol enhances the sensitivity of hepatocellular carcinoma cells to oxaliplatin via metabolic reprogramming. Int J Oncol 2020; 57:733-742. [PMID: 32705170 PMCID: PMC7384842 DOI: 10.3892/ijo.2020.5098] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 06/22/2020] [Indexed: 01/02/2023] Open
Abstract
The Warburg effect is a unique metabolic feature of the majority of tumor cells and is closely related to chemotherapeutic resistance. Pyruvate dehydrogenase kinase 1 (PDK1) is considered a 'switch' that controls the fate of pyruvate in glucose metabolism. However, to date, to the best of our knowledge, there are only a few studies to available which had studied the reduction of chemotherapeutic resistance via the metabolic reprogramming of tumor cells with PDK1 as a target. In the present study, it was found dicoumarol (DIC) reduced the phosphorylation of pyruvate dehydrogenase (PDH) by inhibiting the activity of PDK1, which converted the metabolism of human hepatocellular carcinoma (HCC) cells to oxidative phosphorylation, leading to an increase in mitochondrial reactive oxygen species ROS (mtROS) and a decrease in mitochondrial membrane potential (MMP), thereby increasing the apoptosis induced by oxaliplatin (OXA). Furthermore, the present study elucidated that the targeting of PDK1 may be a potential strategy for targeting metabolism in the chemotherapy of HCC. In addition, DIC as an 'old drug' exhibits novel efficacy, bringing new hope for antitumor therapy.
Collapse
Affiliation(s)
- Huadan Xu
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yichun He
- Department of Neurosurgery, China‑Japan Union Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jiaoyan Ma
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yuanxin Zhao
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yanan Liu
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Liankun Sun
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jing Su
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
17
|
Qin YJ, Lin TY, Lin XL, Liu Y, Zhao WT, Li XY, Lian M, Chen HW, Li YL, Zhang XL, Xiao D, Jia JS, Sun Y. Loss of PDK4 expression promotes proliferation, tumorigenicity, motility and invasion of hepatocellular carcinoma cells. J Cancer 2020; 11:4397-4405. [PMID: 32489458 PMCID: PMC7255379 DOI: 10.7150/jca.43459] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 03/27/2020] [Indexed: 02/07/2023] Open
Abstract
Although the roles and underlying mechanisms of other PDK family members (i.e., PDK1, PDK2 and PDK3) in tumor progression have been extensively investigated and are well understood, the functions and underlying molecular mechanisms of pyruvate dehydrogenase kinase 4 (PDK4) in the tumorigenesis and progression of various cancers [including hepatocellular carcinoma (HCC)] remain largely unknown. In this study, we examined the expression profile of PDK4 in HCC clinical tissue specimens and the roles of PDK4 in the proliferation, tumorigenicity, motility and invasion of HCC cells. The immunohistochemistry (IHC) and quantitative real-time PCR (qRT-PCR) results revealed that PDK4 was significantly downregulated in the cohort of HCC clinical specimens. Additionally, PDK4 protein was found in both the nucleus and cytoplasm of HCC cells based on an immunofluorescence (ICC) assay, and PDK4 protein was also found in the nucleus and cytoplasm of cancer cells contained in HCC clinical specimens based on IHC. The CCK-8 assay and cell colony formation assay demonstrated that stable depletion of endogenous PDK4 by lentivirus-mediated RNA interference (RNAi) markedly promoted the proliferation of HCC cell lines (i.e., BEL-7402 and BEL-7404 cells) in vitro, while PDK4 silencing significantly enhanced the tumorigenic ability of BEL-7404 cells in vivo. In addition to enhance proliferation and tumorigenesis induced by PDK4 silencing, additional studies demonstrated that knockdown of PDK4 led to increase migration and invasion of BEL-7402 and BEL-7404 cells in vitro. Taken together, these findings suggest that the loss of PDK4 expression contributes to HCC malignant progression.
Collapse
Affiliation(s)
- Yu-Juan Qin
- Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou 510515, China
- Department of Radiology, The 5th Affiliated Hospital of Sun Yat-Sen University, Zhuhai 519000, China
| | - Tao-Yan Lin
- Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xiao-Lin Lin
- Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yu Liu
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou 510515, China
| | - Wen-Tao Zhao
- Department of Medical Oncology, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming 650118, China
| | - Xiao-Yan Li
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou 510515, China
| | - Mei Lian
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou 510515, China
| | - Heng-Wei Chen
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou 510515, China
| | - Yong-Long Li
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou 510515, China
| | - Xiao-Ling Zhang
- Department of Physiology, Faculty of Basic Medical Sciences, Guilin Medical University, Guilin 541004, China
| | - Dong Xiao
- Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou 510515, China
| | - Jun-Shuang Jia
- Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yan Sun
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
18
|
Khan MA, Zubair H, Anand S, Srivastava SK, Singh S, Singh AP. Dysregulation of metabolic enzymes in tumor and stromal cells: Role in oncogenesis and therapeutic opportunities. Cancer Lett 2020; 473:176-185. [PMID: 31923436 PMCID: PMC7067140 DOI: 10.1016/j.canlet.2020.01.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/12/2019] [Accepted: 01/03/2020] [Indexed: 01/15/2023]
Abstract
Altered cellular metabolism is a hallmark of cancer. Metabolic rewiring in cancer cells occurs due to the activation of oncogenes, inactivation of tumor suppressor genes, and/or other adaptive changes in cell signaling pathways. Furthermore, altered metabolism is also reported in tumor-corrupted stromal cells as a result of their interaction with cancer cells or due to their adaptation in the dynamic tumor microenvironment. Metabolic alterations are associated with dysregulation of metabolic enzymes and tumor-stromal metabolic crosstalk is vital for the progressive malignant journey of the tumor cells. Therefore, several therapies targeting metabolic enzymes have been evaluated and/or are being investigated in preclinical and clinical studies. In this review, we discuss some important metabolic enzymes that are altered in tumor and/or stromal cells, and focus on their role in supporting tumor growth. Moreover, we also discuss studies carried out in various cancers to target these metabolic abnormalities for therapeutic exploitation.
Collapse
Affiliation(s)
- Mohammad Aslam Khan
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, 36617, USA; Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA
| | - Haseeb Zubair
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, 36617, USA; Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA
| | - Shashi Anand
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, 36617, USA; Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA
| | - Sanjeev Kumar Srivastava
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, 36617, USA; Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA
| | - Seema Singh
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, 36617, USA; Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA; Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL, 36688, USA
| | - Ajay Pratap Singh
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, 36617, USA; Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA; Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL, 36688, USA.
| |
Collapse
|
19
|
Zhao Y, Tran M, Wang L, Shin DJ, Wu J. PDK4-Deficiency Reprograms Intrahepatic Glucose and Lipid Metabolism to Facilitate Liver Regeneration in Mice. Hepatol Commun 2020; 4:504-517. [PMID: 32258946 PMCID: PMC7109344 DOI: 10.1002/hep4.1484] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 01/10/2020] [Indexed: 12/29/2022] Open
Abstract
Liver regeneration requires intrahepatic and extrahepatic metabolic reprogramming to meet the high hepatic bioenergy demand for liver cell repopulation. This study aims to elucidate how pyruvate dehydrogenase kinase 4 (PDK4), a critical regulator of glucose and lipid metabolism, coordinates metabolic regulation with efficient liver growth. We found that hepatic Pdk4 expression was elevated after two-thirds partial hepatectomy (PHx). In Pdk4 -/- PHx mice, the liver/body weight ratio was more rapidly restored, accompanied by more aggressive hepatic DNA replication; however, Pdk4 -/- mice developed more severe hypoglycemia. In Pdk4 -/- PHx livers, the pro-regenerative insulin signaling was potentiated, as demonstrated by early peaking of the phosphorylation of insulin receptor, more remarkable induction of the insulin receptor substrate proteins, IRS1 and IRS2, and more striking activation of Akt. The hepatic up-regulation of CD36 contributed to the enhanced transient regeneration-associated steatosis in Pdk4 -/- PHx mice. Notably, CD36 overexpression in mice promoted the recovery of liver/body weight ratio and elevated intrahepatic adenosine triphosphate after PHx. CD36 expression was transcriptionally suppressed by FOXO1 (forkhead box protein O1), which was stabilized and translocated to the nucleus following AMPK (adenosine monophosphate-activated protein kinase) activation. PHx remarkably induced AMPK activation, which became incompetent to respond in Pdk4 -/- livers. Moreover, we defined that PDK4-regulated AMPK activation directly depended on intracellular adenosine monophosphate in vitro and in regenerative livers. Conclusion: PDK4 inhibition reprograms glucose and lipid metabolism to promote liver regeneration by enhancing hepatic insulin/Akt signaling and activating an AMPK/FOXO1/CD36 regulatory axis of lipid. These findings may lead to potential therapeutic strategies to prevent hepatic insufficiency and liver failure.
Collapse
Affiliation(s)
- Yulan Zhao
- Department of Physiology & Neurobiology University of Connecticut Storrs CT
| | - Melanie Tran
- Department of Physiology & Neurobiology University of Connecticut Storrs CT
| | - Li Wang
- Department of Internal Medicine Section of Digestive Diseases Yale University New Haven CT
| | - Dong-Ju Shin
- Department of Physiology & Neurobiology University of Connecticut Storrs CT
| | - Jianguo Wu
- Department of Physiology & Neurobiology University of Connecticut Storrs CT
| |
Collapse
|
20
|
Atas E, Oberhuber M, Kenner L. The Implications of PDK1-4 on Tumor Energy Metabolism, Aggressiveness and Therapy Resistance. Front Oncol 2020. [PMID: 33384955 DOI: 10.3389/fonc.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2023] Open
Abstract
A metabolic shift from oxidative phosphorylation (OXPHOS) to glycolysis-known as the Warburg effect-is characteristic for many cancers. It gives the cancer cells a survival advantage in the hypoxic tumor microenvironment and protects them from cytotoxic effects of oxidative damage and apoptosis. The main regulators of this metabolic shift are the pyruvate dehydrogenase complex and pyruvate dehydrogenase kinase (PDK) isoforms 1-4. PDK is known to be overexpressed in several cancers and is associated with bad prognosis and therapy resistance. Whereas the expression of PDK1-3 is tissue specific, PDK4 expression is dependent on the energetic state of the whole organism. In contrast to other PDK isoforms, not only oncogenic, but also tumor suppressive functions of PDK4 have been reported. In tumors that profit from high OXPHOS and high de novo fatty acid synthesis, PDK4 can have a protective effect. This is the case for prostate cancer, the most common cancer in men, and makes PDK4 an interesting therapeutic target. While most work is focused on PDK in tumors characterized by high glycolytic activity, little research is devoted to those cases where PDK4 acts protective and is therefore highly needed.
Collapse
Affiliation(s)
- Emine Atas
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Monika Oberhuber
- Department of Pathology, Medical University of Vienna, Vienna, Austria
- Area 'Data & Technologies', CBmed-Center for Biomarker Research in Medicine GmbH, Graz, Austria
| | - Lukas Kenner
- Department of Pathology, Medical University of Vienna, Vienna, Austria
- Area 'Data & Technologies', CBmed-Center for Biomarker Research in Medicine GmbH, Graz, Austria
- Unit of Pathology of Laboratory Animals, University of Veterinary Medicine Vienna, Vienna, Austria
- Christian Doppler Laboratory for Applied Metabolomics (CDL AM), Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
21
|
Woolbright BL, Rajendran G, Harris RA, Taylor JA. Metabolic Flexibility in Cancer: Targeting the Pyruvate Dehydrogenase Kinase:Pyruvate Dehydrogenase Axis. Mol Cancer Ther 2019; 18:1673-1681. [PMID: 31511353 DOI: 10.1158/1535-7163.mct-19-0079] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 05/23/2019] [Accepted: 07/24/2019] [Indexed: 11/16/2022]
Abstract
Cancer cells use alterations of normal metabolic processes to sustain proliferation indefinitely. Transcriptional and posttranscriptional control of the pyruvate dehydrogenase kinase (PDK) family is one way in which cancer cells alter normal pyruvate metabolism to fuel proliferation. PDKs can phosphorylate and inactivate the pyruvate dehydrogenase complex (PDHC), which blocks oxidative metabolism of pyruvate by the mitochondria. This process is thought to enhance cancer cell growth by promoting anabolic pathways. Inhibition of PDKs induces cell death through increased PDH activity and subsequent increases in ROS production. The use of PDK inhibitors has seen widespread success as a potential therapeutic in laboratory models of multiple cancers; however, gaps still exist in our understanding of the biology of PDK regulation and function, especially in the context of individual PDKs. Efforts are currently underway to generate PDK-specific inhibitors and delineate the roles of individual PDK isozymes in specific cancers. The goal of this review is to understand the regulation of the PDK isozyme family, their role in cancer proliferation, and how to target this pathway therapeutically to specifically and effectively reduce cancer growth.
Collapse
Affiliation(s)
| | | | - Robert A Harris
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas
| | - John A Taylor
- Department of Urology, University of Kansas Medical Center, Kansas City, Kansas.
| |
Collapse
|
22
|
Yang Z, Kusumanchi P, Ross RA, Heathers L, Chandler K, Oshodi A, Thoudam T, Li F, Wang L, Liangpunsakul S. Serum Metabolomic Profiling Identifies Key Metabolic Signatures Associated With Pathogenesis of Alcoholic Liver Disease in Humans. Hepatol Commun 2019; 3:542-557. [PMID: 30976744 PMCID: PMC6442705 DOI: 10.1002/hep4.1322] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 01/11/2019] [Indexed: 12/17/2022] Open
Abstract
Alcoholic liver disease (ALD) develops in a subset of heavy drinkers (HDs). The goals of our study were to (1) characterize the global serum metabolomic changes in well-characterized cohorts of controls (Cs), HDs, and those with alcoholic cirrhosis (AC); (2) identify metabolomic signatures as potential diagnostic markers, and (3) determine the trajectory of serum metabolites in response to alcohol abstinence. Serum metabolic profiling was performed in 22 Cs, 147 HDs, and 33 patients with AC using ultraperformance liquid chromatography-tandem mass spectrometry. Hepatic gene expression was conducted in Cs (n = 16) and those with AC (n = 32). We found progressive changes in the quantities of metabolites from heavy drinking to AC. Taurine-conjugated bile acids (taurocholic acid [TCA], 127-fold; taurochenodeoxycholic acid [TCDCA], 131-fold; and tauroursodeoxycholic acid, 56-fold) showed more striking elevations than glycine-conjugated forms (glycocholic acid [GCA], 22-fold; glycochenodeoxycholic acid [GCDCA], 22-fold; and glycoursodeoxycholic acid [GUDCA], 11-fold). This was associated with increased liver cytochrome P450, family 7, subfamily B, member 1 and taurine content (more substrates); the latter was due to dysregulation of homocysteine metabolism. Increased levels of GCDCA, TCDCA, GCA, and TCA positively correlated with disease progression from Child-Pugh A to C and Model for End-Stage Liver Disease scores, whereas GCDCA, GCA, and GUDCA were better predictors of alcohol abstinence. The levels of glucagon-like peptide 1 (GLP-1) and fibroblast growth factor (FGF) 21 but not FGF19 were increased in HDs, and all three were further increased in those with AC. Conclusion: Serum taurine/glycine-conjugated bile acids could serve as noninvasive markers to predict the severity of AC, whereas GLP-1 and FGF21 may indicate a progression from heavy drinking to AC.
Collapse
Affiliation(s)
- Zhihong Yang
- Division of Gastroenterology and Hepatology, Department of MedicineIndiana University School of MedicineIndianapolisIN
| | - Praveen Kusumanchi
- Division of Gastroenterology and Hepatology, Department of MedicineIndiana University School of MedicineIndianapolisIN
| | - Ruth A. Ross
- Division of Gastroenterology and Hepatology, Department of MedicineIndiana University School of MedicineIndianapolisIN
| | - Laura Heathers
- Division of Gastroenterology and Hepatology, Department of MedicineIndiana University School of MedicineIndianapolisIN
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIN
| | - Kristina Chandler
- Division of Gastroenterology and Hepatology, Department of MedicineIndiana University School of MedicineIndianapolisIN
| | - Adepeju Oshodi
- Division of Gastroenterology and Hepatology, Department of MedicineIndiana University School of MedicineIndianapolisIN
| | - Themis Thoudam
- Division of Gastroenterology and Hepatology, Department of MedicineIndiana University School of MedicineIndianapolisIN
- Department of Biomedical ScienceKyungpook National UniversityDaeguSouth Korea
| | - Feng Li
- Department of Molecular and Cellular BiologyBaylor College of MedicineHoustonTX
| | - Li Wang
- Department of Physiology and Neurobiology and the Institute for Systems GenomicsUniversity of ConnecticutStorrsCT
- Veterans Affairs Connecticut Healthcare SystemWest HavenCT
- Department of Internal Medicine, Liver CenterYale UniversityNew HavenCT
| | - Suthat Liangpunsakul
- Division of Gastroenterology and Hepatology, Department of MedicineIndiana University School of MedicineIndianapolisIN
- Department of Biochemistry and Molecular BiologyIndiana University School of MedicineIndianapolisIN
- Roudebush Veterans Administration Medical CenterIndianapolisIN
| |
Collapse
|
23
|
Strowitzki MJ, Radhakrishnan P, Pavicevic S, Scheer J, Kimmer G, Ritter AS, Tuffs C, Volz C, Vondran F, Harnoss JM, Klose J, Schmidt T, Schneider M. High hepatic expression of PDK4 improves survival upon multimodal treatment of colorectal liver metastases. Br J Cancer 2019; 120:675-688. [PMID: 30808993 PMCID: PMC6461828 DOI: 10.1038/s41416-019-0406-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 01/30/2019] [Accepted: 01/31/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Patients with borderline resectable colorectal liver metastases (CRLM) frequently receive neoadjuvant chemotherapy (NC) to reduce tumour burden, thus making surgical resection feasible. Even though NC can induce severe liver injury, most studies investigating tissue-based prognostic markers focus on tumour tissue. Here, we assessed the prognostic significance of pyruvate-dehydrogenase-kinase isoenzyme 4 (PDK4) within liver tissue of patients undergoing surgical resection due to CRLM. METHODS Transcript levels of hypoxia-adaptive genes (such as PDK isoenzymes) were assessed in the tissue of healthy liver, corresponding CRLM, healthy colon mucosa and corresponding tumour. Uni- and multivariate analyses were performed. Responses to chemotherapy upon up- or down-regulation of PDK4 were studied in vitro. RESULTS PDK4 expression within healthy liver tissue was associated with increased overall survival and liver function following surgical resection of CRLM. This association was enhanced in patients with NC. PDK4 expression in CRLM tissue did not correlate with overall survival. Up-regulation of PDK4 increased the resistance of hepatocytes and colon cancer cells against chemotherapy-induced toxicity, whereas knockdown of PDK4 enhanced chemotherapy-associated cell damage. CONCLUSION Our findings suggest that up-regulated PDK4 expression reduces hepatic chemotherapy-induced oxidative stress and is associated with improved postoperative liver function in patients undergoing multimodal treatment and resection of CRLM.
Collapse
Affiliation(s)
- Moritz J Strowitzki
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Praveen Radhakrishnan
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Sandra Pavicevic
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Jana Scheer
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Gwendolyn Kimmer
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Alina S Ritter
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Christopher Tuffs
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Claudia Volz
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Florian Vondran
- Regenerative Medicine and Experimental Surgery (ReMediES), Department of General, Visceral and Transplantation Surgery, Hannover Medical School, Hannover, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Hannover, Germany
| | - Jonathan M Harnoss
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Johannes Klose
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Thomas Schmidt
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Martin Schneider
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
24
|
H19 potentiates let-7 family expression through reducing PTBP1 binding to their precursors in cholestasis. Cell Death Dis 2019; 10:168. [PMID: 30778047 PMCID: PMC6379488 DOI: 10.1038/s41419-019-1423-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 12/27/2018] [Accepted: 01/21/2019] [Indexed: 12/22/2022]
Abstract
Cholestasis induces the hepatic long non-coding RNA H19, which promotes the progression of cholestatic liver fibrosis. However, microRNAs that are dysregulated by H19 during cholestasis remain elusive. Using miRNA-sequencing analysis followed by qPCR validation, we identified marked upregulation of eight members of the let-7 family in cholestatic livers by bile duct ligation (BDL) and H19 overexpression. In particular, the expression of let-7a-1/7d/7f-1 was highly induced in H19-BDL livers but decreased in H19KO-BDL livers. Interestingly, H19 decreased the nuclear let-7 precursors as well as the primary transcripts of let-7a-1/7d/7f-1 levels in BDL mouse livers. Bioinformatics, RNA pull-down, and RNA immunoprecipitation (RIP) assays revealed that the crucial RNA-binding protein polypyrimidine tract-binding protein 1 (PTBP1), an H19 interaction partner, interacted with the precursors of let-7a-1 and let-7d and suppressed their maturation. Both PTBP1 and let-7 expression was differentially regulated by different bile acid species in hepatocyte and cholangiocyte cells. Further, H19 negatively regulated PTBP1's mRNA and protein levels but did not affect its subcellular distribution in BDL mouse livers. Moreover, we found that H19 restrained but PTBP1 facilitated the bioavailability of let-7 miRNAs to their targets. Taken together, this study revealed for the first time that H19 promoted let-7 expression by decreasing PTBP1's expression level and its binding to the let-7 precursors in cholestasis.
Collapse
|
25
|
Yang C, Wang S, Ruan H, Li B, Cheng Z, He J, Zuo Q, Yu C, Wang H, Lv Y, Gu D, Jin G, Yao M, Qin W, Jin H. Downregulation of PDK4 Increases Lipogenesis and Associates with Poor Prognosis in Hepatocellular Carcinoma. J Cancer 2019; 10:918-926. [PMID: 30854098 PMCID: PMC6400816 DOI: 10.7150/jca.27226] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Accepted: 11/09/2018] [Indexed: 12/30/2022] Open
Abstract
Alterations in cellular metabolism are one of the characteristics in cancer. They are not only the result of tumor progression but also the cause of cancer initiation. Pyruvate dehydrogenase kinase 4 (PDK4) is a key metabolic enzyme, which regulates cell metabolism by inhibiting pyruvate dehydrogenase (PDH). However, the function and regulating mechanism of PDK4 in HCC remain unclear. Here, we found that the expression of PDK4 was significantly decreased in HCC tissues, and its downregulation could predict poor prognosis of HCC patients. Silencing PDK4 significantly facilitated proliferation and migration of HCC cells. Knockdown of PDK4 didn't influence the oxidative phosphorylation and glycolysis capacity of HCC cells in vitro. However, knockdown of PDK4 increased expression of key lipogenic enzymes, fatty acid synthase (FASN) and stearoyl-CoA desaturase (SCD), which finally induced lipogenesis. These data suggest that PDK4 inhibits proliferation and migration of HCC cells probably via suppressing lipogenesis.
Collapse
Affiliation(s)
- Chen Yang
- Shanghai Medical College of Fudan University, Shanghai, China.,State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Siying Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haoyu Ruan
- Shanghai Medical College of Fudan University, Shanghai, China
| | - Botai Li
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Zhuoan Cheng
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Jia He
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiaozhu Zuo
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chengtao Yu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Hui Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanyuan Lv
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dishui Gu
- Department of Pathophysiology, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, Guangdong, 523808, China
| | - Guangzhi Jin
- Department of Pathology, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, China
| | - Ming Yao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenxin Qin
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haojie Jin
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
26
|
Wu J, Zhao Y, Park YK, Lee JY, Gao L, Zhao J, Wang L. Loss of PDK4 switches the hepatic NF-κB/TNF pathway from pro-survival to pro-apoptosis. Hepatology 2018; 68:1111-1124. [PMID: 29603325 PMCID: PMC6165716 DOI: 10.1002/hep.29902] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 03/10/2018] [Accepted: 03/23/2018] [Indexed: 12/11/2022]
Abstract
It has been established that nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) members promote survival by upregulating antiapoptotic genes and that genetic and pharmacological inhibition of NF-κB is required for tumor necrosis factor (TNF)-induced hepatocyte apoptosis. In this study, we demonstrate that this pro-survival pathway is switched to pro-apoptosis under pyruvate dehydrogenase kinase 4 (PDK4)-deficient conditions. PDK4-deficiency triggered hepatic apoptosis concomitantly with increased numbers of aberrant mitochondria, reactive oxygen species (ROS) production, sustained c-Jun N-terminal Kinase (JNK) activation, and reduction of glutathione (GSH). Interestingly, PDK4 retained p65 in cytoplasm via a direct protein-protein interaction. Disruption of PDK4-p65 association promoted p65 nuclear translocation. This, in turn, facilitated p65 binding to the TNF promoter to activate TNF-TNFR1 apoptotic pathway. Pdk4-/- livers were sensitized to Jo2 and D-(+)-Galactosamine /Lipopolysaccharide (GalN/LPS)-mediated apoptotic injury which was prevented by the inhibition of p65 or TNFR1. The pro-survival activity of TNF was shifted, which was switched to a pro-apoptotic activity in Pdk4-/- hepatocytes as a result of impaired activation of pro-survival NF-κB targets. Conclusion: PDK4 is indispensable to dictate the fate of TNF/NF-κB-mediated hepatocyte apoptosis. (Hepatology 2018).
Collapse
Affiliation(s)
- Jianguo Wu
- Department of Physiology and Neurobiology, Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269,Corresponding author: Jianguo Wu (), 75 North Eagleville Rd., U3156, Storrs, CT 06269. Tel: 860-486-0857; Fax: 860-486-3303
| | - Yulan Zhao
- Department of Physiology and Neurobiology, Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269
| | - Young-Ki Park
- Department of Nutritional Sciences, Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269
| | - Ji-Young Lee
- Department of Nutritional Sciences, Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269
| | - Ling Gao
- Department of Endocrinology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong, 250021, China,Shandong Clinical Medical Center of Endocrinology and Metabolism, Jinan, Shandong, 250021, China,Institute of Endocrinology and metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong, 250021, China
| | - Jiajun Zhao
- Department of Endocrinology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong, 250021, China,Shandong Clinical Medical Center of Endocrinology and Metabolism, Jinan, Shandong, 250021, China,Institute of Endocrinology and metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong, 250021, China
| | - Li Wang
- Department of Physiology and Neurobiology, Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269,Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516,Department of Internal Medicine, Section of Digestive Diseases, Yale University, New Haven, CT 06520
| |
Collapse
|
27
|
The miR-15b-5p/PDK4 axis regulates osteosarcoma proliferation through modulation of the Warburg effect. Biochem Biophys Res Commun 2018; 503:2749-2757. [PMID: 30093112 DOI: 10.1016/j.bbrc.2018.08.035] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 08/04/2018] [Indexed: 02/08/2023]
Abstract
Blocking aerobic glycolysis has been proposed as an attractive therapeutic strategy for impairing the proliferation of cancer cells. However, the underlying mechanisms are poorly understood. Here, we show that miR-15b-5p was downregulated in osteosarcoma (OS) and that lower expression of miR-15b-5p promoted proliferation and contributed to the Warburg effect in OS cells. Mechanistically, miR-15b-5p acted as a tumor suppressor in OS by directly targeting pyruvate dehydrogenase kinase-4 and inhibiting its expression. These results reveal a previously unknown function of miR-15b-5p in OS, which is associated with metabolic alterations that promote cancer progression. miR-15b-5p may play an essential role in the molecular therapy of patients with OS.
Collapse
|
28
|
Choiniere J, Lin MJ, Wang L, Wu J. Deficiency of pyruvate dehydrogenase kinase 4 sensitizes mouse liver to diethylnitrosamine and arsenic toxicity through inducing apoptosis. LIVER RESEARCH 2018; 2:100-107. [PMID: 31815032 PMCID: PMC6896988 DOI: 10.1016/j.livres.2018.05.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIM Pyruvate dehydrogenase kinase 4 (PDK4) is a metabolism switch that regulates glucose oxidation and the tricarboxylic acid cycle (TCA cycle) in the mitochondria. Liver detoxifies xenobiotics and is constantly challenged by various injuries. This study aims at understanding how the loss of the metabolism regulator PDK4 contributes to liver injuries. METHODS Wild-type (WT) and Pdk4 knockout (Pdk4 -/-) mice of different ages were examined for spontaneous hepatic apoptosis. Juvenile or adult mice of two genotypes were insulted by diethylnitrosamine (DEN), arsenic, galactosamine (GalN)/lipopolysaccharide (LPS), anti-CD95 (Jo2) antibody or carbon tetrachloride (CCl4). Liver injury was monitored by blood biochemistry test. Apoptosis was determined by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining, poly (ADP-ribose) polymerase (PARP) cleavage, and caspase activity assay. Inflammatory response was determined by nuclear factor (NF)-κB activation and the activation of NF-κB target genes. Primary hepatocytes were isolated and cell viability was evaluated by MTS assay. RESULTS We showed that systematic Pdk4 -/- in mice resulted in age-dependent spontaneous hepatic apoptosis. PDK4-deficiency increased the toxicity of DEN in juvenile mice, which correlated with a lethal consequence and massive hepatic apoptosis. Similarly, chronic arsenic administration induced more severe hepatic apoptosis in Pdk4 -/- mice compared to WT control mice. An aggravated hepatic NF-κB mediated-inflammatory response was observed in Pdk4 -/- mice livers. In vitro, Pdk4-deficient primary hepatocytes were more vulnerable to DEN and arsenic challenges and displayed higher caspase activity than wild type cells. Notably, hepatic PDK4 mRNA level was remarkably reduced during acute liver failure induced by GalN/LPS or Jo2 antibody. The diminished PDK4 expression was also observed in CCl4-induced acute liver injury. CONCLUSIONS PDK4 may contribute to the protection from apoptotic injury in mouse liver.
Collapse
Affiliation(s)
- Jonathan Choiniere
- Department of Physiology and Neurobiology, Institute for Systems Genomics, University of Connecticut, Storrs, CT, USA
| | - Matthew Junda Lin
- Department of Physiology and Neurobiology, Institute for Systems Genomics, University of Connecticut, Storrs, CT, USA
| | - Li Wang
- Department of Physiology and Neurobiology, Institute for Systems Genomics, University of Connecticut, Storrs, CT, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
- Department of Internal Medicine, Section of Digestive Diseases, Yale University, New Haven, CT, USA
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jianguo Wu
- Department of Physiology and Neurobiology, Institute for Systems Genomics, University of Connecticut, Storrs, CT, USA
| |
Collapse
|
29
|
Zhang Y, Zhao Y, Wu J, Liangpunsakul S, Niu J, Wang L. MicroRNA-26-5p functions as a new inhibitor of hepatoblastoma by repressing lin-28 homolog B and aurora kinase a expression. Hepatol Commun 2018; 2:861-871. [PMID: 30027143 PMCID: PMC6049067 DOI: 10.1002/hep4.1185] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 03/10/2018] [Accepted: 03/15/2018] [Indexed: 12/16/2022] Open
Abstract
Hepatoblastoma (HB) is the most common liver tumor in children. Despite recent improvements in treatment strategies, the survival of children with hepatoblastoma remains poor. In this study, we identified a novel role of microRNA‐26a‐5p (miR‐26a‐5p), lin‐28 homolog B (LIN28B), Ras‐related nuclear protein (RAN), and aurora kinase A (AURKA) in HB. The expression of LIN28B, RAN, and AURKA was significantly up‐regulated in human HB livers and cell lines. Knockdown of LIN28B and RAN by small interfering RNAs inhibited HB tumor cell proliferation and foci formation. We also elucidated miR‐26a‐5p‐mediated translational inhibition of LIN28B and AURKA in HB. Overexpression of miR‐26a‐5p markedly decreased LIN28B and AURKA 3′‐untranslated region activities and protein expression and repressed HB cell proliferation and colony formation. In contrast, re‐expression of LIN28B and AURKA rescued miR‐26a‐5p‐mediated suppression of HB cell growth and clonality. Importantly, a decreased miR‐26a‐5p expression correlated with the poor outcome of patients with HB. Conclusion: miR‐26a‐5p is a newly identified repressor of HB growth through its inhibition of the oncogenic LIN28B–RAN–AURKA pathway. (Hepatology Communications 2018;2:481‐491)
Collapse
Affiliation(s)
- Yutong Zhang
- Department of Physiology and Neurobiology and Institute for Systems Genomics University of Connecticut Storrs CT.,Department of Pediatric Oncology The First Hospital of Jilin University Changchun China
| | - Yulan Zhao
- Department of Physiology and Neurobiology and Institute for Systems Genomics University of Connecticut Storrs CT
| | - Jianguo Wu
- Department of Physiology and Neurobiology and Institute for Systems Genomics University of Connecticut Storrs CT
| | - Suthat Liangpunsakul
- Division of Gastroenterology and Hepatology, Department of Medicine Indiana University School of Medicine Indianapolis IN.,Department of Biochemistry and Molecular Biology Indiana University School of Medicine Indianapolis IN.,Roudebush Veterans Administration Medical Center Indianapolis IN
| | - Junqi Niu
- Department of Hepatology The First Hospital of Jilin University Changchun China
| | - Li Wang
- Department of Physiology and Neurobiology and Institute for Systems Genomics University of Connecticut Storrs CT.,Department of Internal Medicine Section of Digestive Diseases, Yale University New Haven CT.,Veterans Affairs Connecticut Healthcare System West Haven CT
| |
Collapse
|