1
|
Pant BD, Ahuja A, Roychowdhury S, Shrestha D, Cross E, Wang Y, Dwyer C, Paxitzis A, Jeng M, Dudekonda S, Scheraga R, Vachharajani V. Mitoquinol improves phagocytosis and glycolysis in ethanol-exposed macrophages via HIF-1α-PFKP axis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf078. [PMID: 40356076 DOI: 10.1093/jimmun/vkaf078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 03/01/2025] [Indexed: 05/15/2025]
Abstract
Alcohol use disorder increases sepsis mortality. Acute ethanol exposure impairs pathogen clearance in the macrophages via dampened glycolysis and phagocytosis, exaggerates oxidative stress, and regulates the function of the hypoxia-regulating factor 1α (HIF-1α), a master regulator of glycolysis. Decreased expression of the platelet isoform of phosphofructokinase (PFKP), a key glycolytic enzyme, in ethanol-exposed macrophages, is reported. However, transcriptional regulation of PFKP with ethanol exposure is unclear. We hypothesized that acute ethanol exposure-induced oxidative stress dampens macrophage phagocytosis and glycolysis via the HIF-1α-PFKP axis. In ethanol-exposed mouse bone marrow-derived macrophages with lipopolysaccharide stimulation, we studied (i) reactive oxygen species (ROS), phagocytosis, glycolysis, PFKP, and HIF-1α expressions ± ethanol exposure; (ii) the role of HIF-1α in transcriptionally controlling PFKP messenger RNA by chromatin immunoprecipitation-quantitative polymerase chain reaction technique; and (iii) the effect of mitoquinol (MitoQ), a mitochondria-specific antioxidant, on HIF-1α function, glycolysis, phagocytosis, and pathogen clearance in ethanol-exposed macrophages. Last, we examined the effect of MitoQ on 7-d survival in alcohol vs. vehicle-drinking mice with cecal slurry-induced sepsis. In ethanol-exposed and lipopolysaccharide-stimulated macrophages, we found that (i) excessive total and mitochondrial ROS production and dampened phagocytosis, glycolysis, and PFKP expression; (ii) dysfunctional HIF-1α downregulates PFKP transcription; (iii) MitoQ restrains ROS production, restores HIF-1α function, and improves glycolysis and phagocytosis via preserved PFKP messenger RNA and protein expression; and (iv) MitoQ treatment improves survival and pathogen clearance in ethanol with sepsis mice. In conclusion, we found that the HIF-1α-PFKP axis regulates glycolysis and phagocytosis in ethanol-exposed macrophages and is a potential therapeutic target in ethanol with sepsis.
Collapse
Affiliation(s)
- Bishnu D Pant
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH, United States
| | - Akash Ahuja
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH, United States
| | - Sanjoy Roychowdhury
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH, United States
| | - Deepmala Shrestha
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH, United States
| | - Emily Cross
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH, United States
| | - Yuxin Wang
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH, United States
| | - Christian Dwyer
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH, United States
| | - Alexandra Paxitzis
- Pulmonary and Critical Care Medicine, Integrated Hospital care Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Margaret Jeng
- Pulmonary and Critical Care Medicine, Integrated Hospital care Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Sudhir Dudekonda
- Pulmonary and Critical Care Medicine, Integrated Hospital care Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Rachel Scheraga
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH, United States
- Pulmonary and Critical Care Medicine, Integrated Hospital care Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Vidula Vachharajani
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH, United States
- Pulmonary and Critical Care Medicine, Integrated Hospital care Institute, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
2
|
Jiang Z, Liu T, Wang Y, Li J, Guo L. Effect of lncRNA XIST on acute myeloid leukemia cells via miR-142-5p-PFKP axis. Hematology 2024; 29:2306444. [PMID: 38305210 DOI: 10.1080/16078454.2024.2306444] [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] [Indexed: 02/03/2024] Open
Abstract
Acute myeloid leukemia (AML) is the common blood cancer in hematopoietic system-related diseases and has a poor prognosis. Studies have shown that long non-coding RNAs (lncRNAs) are closely related to the pathogenesis of a variety of diseases, including AML. However, the specific molecular mechanism remains unclear. Hence, the objective of this study was to investigate the effect and mechanism of lncRNA X inactive specific transcript (lncRNA XIST) on AML. To achieve our objective, some tests were performed. Quantitative real-time polymerase chain reaction (qRT-PCR) was utilized to detect the expression of lncRNA XIST, miR-142-5p and the platelet isoform of phosphofructokinase (PFKP). The targeting relationship between miR-142-5p and lncRNA XIST and PFKP was verified by Pearson correlation analysis, dual-luciferase reporter assay, and pull-down assay. Functional experiments were used to analyze the effect and mechanism of action of knocking down lncRNA XIST on THP-1 and U937 cells. Compared with bone marrow cells, lncRNA XIST and PFKP expression levels were up-regulated and miR-142-5p expression levels were down-regulated in AML. Further analysis revealed that lncRNA XIST targeted and bound to miR-142-5p, and PFKP was a target gene of miR-142-5p. Knockdown of lncRNA XIST significantly promoted miR-142-5p expression to down-regulate PFKP in THP-1 and U937 cells, while the cell proliferation, cell viability, and cell cycle arrest were inhibited and apoptosis was increased. Knockdown of miR-142-5p reversed the functional impact of lncRNA XIST knockdown on AML cells. In conclusion, down-regulation of lncRNA XIST can affect the progression of AML by regulating miR-142-5p.
Collapse
Affiliation(s)
- Zhaozhi Jiang
- Blood Transfusion Department, Affiliated Hospital of Jilin Medical University, Jilin, China
| | - Tingting Liu
- Pathology Department, Affiliated Hospital of Jilin Medical University, Jilin, China
| | - Youhong Wang
- Blood Transfusion Department, Affiliated Hospital of Jilin Medical University, Jilin, China
| | - Jiao Li
- Blood Transfusion Department, Affiliated Hospital of Jilin Medical University, Jilin, China
| | - Lusheng Guo
- Blood Transfusion Department, Affiliated Hospital of Jilin Medical University, Jilin, China
| |
Collapse
|
3
|
Wang W, Dai R, Cheng M, Chen Y, Gao Y, Hong X, Zhang W, Wang Y, Zhang L. Metabolic reprogramming and renal fibrosis: what role might Chinese medicine play? Chin Med 2024; 19:148. [PMID: 39465434 PMCID: PMC11514863 DOI: 10.1186/s13020-024-01004-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: 07/11/2024] [Accepted: 09/15/2024] [Indexed: 10/29/2024] Open
Abstract
Metabolic reprogramming is a pivotal biological process in which cellular metabolic patterns change to meet the energy demands of increased cell growth and proliferation. In this review, we explore metabolic reprogramming and its impact on fibrotic diseases, providing a detailed overview of the key processes involved in the metabolic reprogramming of renal fibrosis, including fatty acid decomposition and synthesis, glycolysis, and amino acid catabolism. In addition, we report that Chinese medicine ameliorates renal inflammation, oxidative stress, and apoptosis in chronic kidney disease by regulating metabolic processes, thereby inhibiting renal fibrosis. Furthermore, we reveal that multiple targets and signaling pathways contribute to the metabolic regulatory effects of Chinese medicine. In summary, this review aims to elucidate the mechanisms by which Chinese medicine inhibits renal fibrosis through the remodeling of renal cell metabolic processes, with the goal of discovering new therapeutic drugs for treating renal fibrosis.
Collapse
Affiliation(s)
- Weili Wang
- First Clinical Medical College, Anhui University of Chinese Medicine, Hefei, China
| | - Rong Dai
- Department of Nephrology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Meishan Road 117, Shushang District, Hefei, 230031, China
| | - Meng Cheng
- Department of Nephrology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Meishan Road 117, Shushang District, Hefei, 230031, China
| | - Yizhen Chen
- First Clinical Medical College, Anhui University of Chinese Medicine, Hefei, China
| | - Yilin Gao
- First Clinical Medical College, Anhui University of Chinese Medicine, Hefei, China
| | - Xin Hong
- First Clinical Medical College, Anhui University of Chinese Medicine, Hefei, China
| | - Wei Zhang
- First Clinical Medical College, Anhui University of Chinese Medicine, Hefei, China
| | - Yiping Wang
- Department of Nephrology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Meishan Road 117, Shushang District, Hefei, 230031, China.
| | - Lei Zhang
- Department of Nephrology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Meishan Road 117, Shushang District, Hefei, 230031, China.
| |
Collapse
|
4
|
Tura A, Herfs V, Maaßen T, Zuo H, Vardanyan S, Prasuhn M, Ranjbar M, Kakkassery V, Grisanti S. Quercetin Impairs the Growth of Uveal Melanoma Cells by Interfering with Glucose Uptake and Metabolism. Int J Mol Sci 2024; 25:4292. [PMID: 38673877 PMCID: PMC11049862 DOI: 10.3390/ijms25084292] [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: 03/13/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Monosomy 3 in uveal melanoma (UM) increases the risk of lethal metastases, mainly in the liver, which serves as the major site for the storage of excessive glucose and the metabolization of the dietary flavonoid quercetin. Although primary UMs with monosomy 3 exhibit a higher potential for basal glucose uptake, it remains unknown as to whether glycolytic capacity is altered in such tumors. Herein, we initially analyzed the expression of n = 151 genes involved in glycolysis and its interconnected branch, the "pentose phosphate pathway (PPP)", in the UM cohort of The Cancer Genome Atlas Study and validated the differentially expressed genes in two independent cohorts. We also evaluated the effects of quercetin on the growth, survival, and glucose metabolism of the UM cell line 92.1. The rate-limiting glycolytic enzyme PFKP was overexpressed whereas the ZBTB20 gene (locus: 3q13.31) was downregulated in the patients with metastases in all cohorts. Quercetin was able to impair proliferation, viability, glucose uptake, glycolysis, ATP synthesis, and PPP rate-limiting enzyme activity while increasing oxidative stress. UMs with monosomy 3 display a stronger potential to utilize glucose for the generation of energy and biomass. Quercetin can prevent the growth of UM cells by interfering with glucose metabolism.
Collapse
Affiliation(s)
- Aysegül Tura
- Department of Ophthalmology, University of Lübeck, Ratzeburger Allee 160, 23562 Luebeck, Germany; (V.H.); (T.M.); (H.Z.); (S.V.); (M.P.); (V.K.); (S.G.)
| | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Coffey NJ, Simon MC. Metabolic alterations in hereditary and sporadic renal cell carcinoma. Nat Rev Nephrol 2024; 20:233-250. [PMID: 38253811 PMCID: PMC11165401 DOI: 10.1038/s41581-023-00800-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2023] [Indexed: 01/24/2024]
Abstract
Kidney cancer is the seventh leading cause of cancer in the world, and its incidence is on the rise. Renal cell carcinoma (RCC) is the most common form and is a heterogeneous disease comprising three major subtypes that vary in their histology, clinical course and driver mutations. These subtypes include clear cell RCC, papillary RCC and chromophobe RCC. Molecular analyses of hereditary and sporadic forms of RCC have revealed that this complex and deadly disease is characterized by metabolic pathway alterations in cancer cells that lead to deregulated oxygen and nutrient sensing, as well as impaired tricarboxylic acid cycle activity. These metabolic changes facilitate tumour growth and survival. Specifically, studies of the metabolic features of RCC have led to the discovery of oncometabolites - fumarate and succinate - that can promote tumorigenesis, moonlighting functions of enzymes, and substrate auxotrophy owing to the disruption of pathways that enable the production of arginine and cholesterol. These metabolic alterations within RCC can be exploited to identify new therapeutic targets and interventions, in combination with novel approaches that minimize the systemic toxicity of metabolic inhibitors and reduce the risk of drug resistance owing to metabolic plasticity.
Collapse
Affiliation(s)
- Nathan J Coffey
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - M Celeste Simon
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
6
|
Wang H, Penaloza T, Manea AJ, Gao X. PFKP: More than phosphofructokinase. Adv Cancer Res 2023; 160:1-15. [PMID: 37704285 PMCID: PMC12125951 DOI: 10.1016/bs.acr.2023.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/01/2023]
Abstract
Phosphofructokinase (PFK) is one of the key enzymes that functions in glycolysis. Studies show that PFKP regulates cell proliferation, apoptosis, autophagy, cell migration/metastasis, and stemness through glycolysis and glycolysis-independent functions. PFKP performs its function not only in the cytoplasm, but also at the cell membrane, on the mitochondria, at the lysosomal membrane, and in the nucleus. The functions of PFKP are extensively studied in cancer cells. PFKP is also highly expressed in certain immune cells; nevertheless, the study of the PFKP's role in immune cells is limited. In this review, we summarize how the expression and activity of PFKP are regulated in cancer cells. PFKP may be applied as a prognostic marker due to its overexpression and significant functions in cancer cells. As such, specifically targeting/inhibiting PFKP may be a critical and promising strategy for cancer therapy.
Collapse
Affiliation(s)
- Haizhen Wang
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States.
| | - Tiffany Penaloza
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States
| | - Amanda J Manea
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States
| | - Xueliang Gao
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States.
| |
Collapse
|
7
|
Lu Y, Liu S, Sun Y, Zhao B, Xu D. Identification of key genes in hepatocellular carcinoma associated with exposure to TCDD and α-endosulfan by WGCNA. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 252:114595. [PMID: 36753968 DOI: 10.1016/j.ecoenv.2023.114595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 01/14/2023] [Accepted: 01/30/2023] [Indexed: 06/18/2023]
Abstract
2,3,7,8-tet-rachlorodibenzo-p-dioxin (TCDD) and α-endosulfan are two typical persistent organic pollutants (POPs), both of which accumulate in the liver and have potential carcinogenic hepatic effects. The underlying molecular mechanisms of pathogenesis of hepatocellular carcinoma (HCC) remain elusive when exposure to POPs. The aim of this study is to explore the key genes involved in HCC when exposure to TCDD and α-endosulfan by weighted gene co-expression network analysis (WGCNA). First, we performed co-expressed analysis on HCC and normal condition, based on WGCNA. In results, seven co-expressed modules were identified from 56 human liver samples, and the brown module correlated with five stages of HCC. Subsequently, we predicted that human five liver diseases were associated with exposure to TCDD and/or α-endosulfan by Nextbio analysis. Functional enrichment analysis showed that the brown module enriched in oxidation-reduction process, DNA replication, oxidoreductase activity and aging, which were the same as the results when exposure to the mixture of TCDD and α-endosulfan. Lastly, based on the protein-protein interaction network, we identified three novel genes including HK2, EXO1 and PFKP as key genes in HCC associated with exposure to TCDD and α-endosulfan mixture. In addition, survival analysis of key genes in Kaplan-Meier plotter demonstrated that aberrant expression levels of all the three key genes were associated with poor prognosis of HCC. Finally, Western blot analysis confirmed that protein expression levels of PFKP and HK2 in the three exposed groups were significantly elevated, while EXO1 were significantly upregulated when exposure to TCDD and α-endosulfan mixture in HepaRG cells. This study provides a new perspective to the understanding of the genetic mechanism of HCC when exposure to POPs.
Collapse
Affiliation(s)
- Yanyuan Lu
- Institute of Environmental Systems Biology, Environment Science and Engineering College, Dalian Maritime University, Linghai Road 1, Dalian 116026, China
| | - Shiqi Liu
- Institute of Environmental Systems Biology, Environment Science and Engineering College, Dalian Maritime University, Linghai Road 1, Dalian 116026, China
| | - Yeqing Sun
- Institute of Environmental Systems Biology, Environment Science and Engineering College, Dalian Maritime University, Linghai Road 1, Dalian 116026, China
| | - Bin Zhao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Shuangqing Road 18, Beijing 100085, China
| | - Dan Xu
- Institute of Environmental Systems Biology, Environment Science and Engineering College, Dalian Maritime University, Linghai Road 1, Dalian 116026, China.
| |
Collapse
|
8
|
Chen C, Zhang X. Glycolysis regulator PFKP induces human melanoma cell proliferation and tumor growth. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2023:10.1007/s12094-023-03096-7. [PMID: 36792847 DOI: 10.1007/s12094-023-03096-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 01/18/2023] [Indexed: 02/17/2023]
Abstract
PURPOSE Cutaneous melanoma is an aggressive and deadly cancer resulting from malignant transformation of cells involved in skin pigmentation. Glycolysis is widely implicated in cancer progression, but its precise role in melanoma has not been extensively studied. Here, we investigated the role of the glycolysis regulator phosphofructokinase 1 platelet isoform (PFKP) in melanoma progression. METHODS PFKP expression in human melanoma tissues was analyzed by immunohistochemistry. Knockdown of PFKP by siRNA and overexpression of PFKP were performed to evaluate its functions in vitro. CCK-8 assay was used to assess cell proliferation. Glycolytic activity was determined via measurement of extracellular acidification rate (ECAR), lactic acid level, and ATP content. A tumor xenograft model was used to test the function of PFKP in vivo. RESULTS PFKP upregulation was observed in human melanoma tissues and correlated with poor patient survival. Knockdown of PFKP in human melanoma cells suppressed cell proliferation and reduced ECAR, ATP levels, and lactic acid levels, while overexpression of PFKP displayed the opposite effects. In vivo, knockdown of PFKP in melanoma cells markedly reduced tumorigenesis. Inhibitory effects on cell proliferation, glycolysis, and tumorigenesis due to PFKP knockdown were further augmented upon treatment with the glycolysis inhibitor 2-deoxy-D-glucose (2-DG). CONCLUSION Collectively, these results indicate that PFKP expression in melanoma cells increases proliferation and glycolytic activity in vitro and promotes tumorigenesis in vivo, suggesting that suppression of PKFP and inhibition of glycolysis may potently suppress melanoma progression.
Collapse
Affiliation(s)
- Cheng Chen
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Fenglin Rd No.180, Shanghai, 200032, China
| | - Xuejun Zhang
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Fenglin Rd No.180, Shanghai, 200032, China.
| |
Collapse
|
9
|
Gandhirajan A, Roychowdhury S, Kibler C, Cross E, Abraham S, Bellar A, Nagy LE, Scheraga RG, Vachharajani V. SIRT2-PFKP interaction dysregulates phagocytosis in macrophages with acute ethanol-exposure. Front Immunol 2023; 13:1079962. [PMID: 36865524 PMCID: PMC9972587 DOI: 10.3389/fimmu.2022.1079962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/27/2022] [Indexed: 01/28/2023] Open
Abstract
Alcohol abuse, reported by 1/8th critically ill patients, is an independent risk factor for death in sepsis. Sepsis kills over 270,000 patients/year in the US. We reported that the ethanol-exposure suppresses innate-immune response, pathogen clearance, and decreases survival in sepsis-mice via sirtuin 2 (SIRT2). SIRT2 is an NAD+-dependent histone-deacetylase with anti-inflammatory properties. We hypothesized that in ethanol-exposed macrophages, SIRT2 suppresses phagocytosis and pathogen clearance by regulating glycolysis. Immune cells use glycolysis to fuel increased metabolic and energy demand of phagocytosis. Using ethanol-exposed mouse bone marrow- and human blood monocyte-derived macrophages, we found that SIRT2 mutes glycolysis via deacetylating key glycolysis regulating enzyme phosphofructokinase-platelet isoform (PFKP), at mouse lysine 394 (mK394, human: hK395). Acetylation of PFKP at mK394 (hK395) is crucial for PFKP function as a glycolysis regulating enzyme. The PFKP also facilitates phosphorylation and activation of autophagy related protein 4B (Atg4B). Atg4B activates microtubule associated protein 1 light chain-3B (LC3). LC3 is a driver of a subset of phagocytosis, the LC3-associated phagocytosis (LAP), which is crucial for segregation and enhanced clearance of pathogens, in sepsis. We found that in ethanol-exposed cells, the SIRT2-PFKP interaction leads to decreased Atg4B-phosphorylation, decreased LC3 activation, repressed phagocytosis and LAP. Genetic deficiency or pharmacological inhibition of SIRT2 reverse PFKP-deacetylation, suppressed LC3-activation and phagocytosis including LAP, in ethanol-exposed macrophages to improve bacterial clearance and survival in ethanol with sepsis mice.
Collapse
Affiliation(s)
- Anugraha Gandhirajan
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH, United States
| | - Sanjoy Roychowdhury
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH, United States
| | - Christopher Kibler
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH, United States
| | - Emily Cross
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH, United States
| | - Susamma Abraham
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH, United States
| | - Annett Bellar
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH, United States
| | - Laura E. Nagy
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH, United States
| | - Rachel Greenberg Scheraga
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH, United States
- Department of Critical Care Medicine, Respiratory Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Vidula Vachharajani
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH, United States
- Department of Critical Care Medicine, Respiratory Institute, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
10
|
Kobayashi H, Takase S, Nishimura H, Matsumoto K, Harada H, Yoshida M. RNAi screening reveals a synthetic chemical-genetic interaction between ATP synthase and PFK1 in cancer cells. Cancer Sci 2023; 114:1663-1671. [PMID: 36601784 PMCID: PMC10067418 DOI: 10.1111/cas.15713] [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: 11/20/2022] [Revised: 12/25/2022] [Accepted: 12/27/2022] [Indexed: 01/06/2023] Open
Abstract
To meet cellular bioenergetic and biosynthetic demands, cancer cells remodel their metabolism to increase glycolytic flux, a phenomenon known as the Warburg effect and believed to contribute to cancer malignancy. Among glycolytic enzymes, phosphofructokinase-1 (PFK1) has been shown to act as a rate-limiting enzyme and to facilitate the Warburg effect in cancer cells. In this study, however, we found that decreased PFK1 activity did not affect cell survival or proliferation in cancer cells. This raised a question regarding the importance of PFK1 in malignancy. To gain insights into the role of PFK1 in cancer metabolism and the possibility of adopting it as a novel anticancer therapeutic target, we screened for genes that caused lethality when they were knocked down in the presence of tryptolinamide (TLAM), a PFK1 inhibitor. The screen revealed a synthetic chemical-genetic interaction between genes encoding subunits of ATP synthase (complex V) and TLAM. Indeed, after TLAM treatment, the sensitivity of HeLa cells to oligomycin A (OMA), an ATP synthase inhibitor, was 13,000 times higher than that of untreated cells. Furthermore, this sensitivity potentiation by TLAM treatment was recapitulated by genetic mutations of PFK1. By contrast, TLAM did not potentiate the sensitivity of normal fibroblast cell lines to OMA, possibly due to their reduced energy demands compared to cancer cells. We also showed that the PFK1-mediated glycolytic pathway can act as an energy reservoir. Selective potentiation of the efficacy of ATP synthase inhibitors by PFK1 inhibition may serve as a foundation for novel anticancer therapeutic strategies.
Collapse
Affiliation(s)
- Hiroki Kobayashi
- Seed Compounds Exploratory Unit for Drug Discovery Platform, RIKEN Center for Sustainable Resource Science, Saitama, Japan.,Laboratory of Oncology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Shohei Takase
- Chemical Genomics Research Group, RIKEN Center for Sustainable Resource Science, Saitama, Japan
| | - Haruna Nishimura
- Seed Compounds Exploratory Unit for Drug Discovery Platform, RIKEN Center for Sustainable Resource Science, Saitama, Japan
| | - Ken Matsumoto
- Seed Compounds Exploratory Unit for Drug Discovery Platform, RIKEN Center for Sustainable Resource Science, Saitama, Japan.,Chemical Genomics Research Group, RIKEN Center for Sustainable Resource Science, Saitama, Japan
| | - Hironori Harada
- Laboratory of Oncology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Minoru Yoshida
- Seed Compounds Exploratory Unit for Drug Discovery Platform, RIKEN Center for Sustainable Resource Science, Saitama, Japan.,Chemical Genomics Research Group, RIKEN Center for Sustainable Resource Science, Saitama, Japan.,Department of Biotechnology, Graduate School of Agricultural Life Sciences, The University of Tokyo, Tokyo, Japan.,Collaborative Research Institute for Innovative Microbiology, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
11
|
Lu TJ, Yang YF, Cheng CF, Tu YT, Chen YR, Lee MC, Tsai KW. Phosphofructokinase Platelet Overexpression Accelerated Colorectal Cancer Cell Growth and Motility. J Cancer 2023; 14:943-951. [PMID: 37151384 PMCID: PMC10158518 DOI: 10.7150/jca.82738] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 03/25/2023] [Indexed: 05/09/2023] Open
Abstract
Background: Glycolysis is a glucose metabolism pathway that generates the high-energy compound adenosine triphosphate, which supports cancer cell growth. Phosphofructokinase platelet (PFKP) plays a crucial role in glycolysis regulation and is involved in human cancer progression. However, the biological function of PFKP remains unclear in colorectal cancer (CRC). Methods: We analyzed the expression levels of PFKF in colon cancer cells and clinical samples using real-time PCR and western blot techniques. To determine the clinical significance of PFKP expression in colorectal cancer (CRC), we analyzed public databases. In addition, we conducted in vitro assays to investigate the effects of PFKP on cell growth, cell cycle, and motility. Results: An analysis by the Cancer Genome Atlas database revealed that PFKP was significantly overexpressed in CRC. We examined the levels of PFKP mRNA and protein, revealing that PFKP expression was significantly increased in CRC. The results of the univariate Cox regression analysis showed that high PFKP expression was linked to worse disease-specific survival (DSS) and overall survival (OS) [DSS: crude hazard ratio (CHR) = 1.84, 95% confidence interval (CI): 1.01-3.36, p = 0.047; OS: CHR=1.91, 95% CI: 1.06-3.43, p = 0.031]. Multivariate Cox regression analysis revealed that high PFKP expression was an independent prognostic biomarker for the DSS and OS of patients with CRC (DSS: adjusted HR = 2.07, 95% CI: 1.13-3.79, p = 0.018; AHR = 2.34, 95% CI: 1.29-4.25, p = 0.005). PFKP knockdown reduced the proliferation, colony formation, and invasion of CRC cells. In addition, the knockdown induced cell cycle arrest at the G0/G1 phase by impairing cell cycle-related protein expression. Conclusion: Overexpression of PFKP contributes to the growth and invasion of CRC by regulating cell cycle progression. PFKP expression can serve as a valuable molecular biomarker for cancer prognosis and a potential therapeutic target for treating CRC.
Collapse
Affiliation(s)
- Tzung-Ju Lu
- Division of Colon and Rectal Surgery, Department of Surgery, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
| | - Yi-Fen Yang
- Pulmonary function Laboratory, Division of Pulmonary Medicine, Kaohsiung Medical University Chung-Ho Memorial Hospital
| | - Ching-Feng Cheng
- Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taipei, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Department of Pediatrics, Tzu Chi University, Hualien, Taiwan
| | - Ya-Ting Tu
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| | - Yi-Ru Chen
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| | - Ming-Cheng Lee
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| | - Kuo-Wang Tsai
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
- Department of Nursing, Cardinal Tien Junior College of Healthcare and Management, Taiwan
- ✉ Corresponding author: Kuo-Wang Tsai, Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan, R. O. C. E-Mail:
| |
Collapse
|
12
|
Lackner M, Neef SK, Winter S, Beer-Hammer S, Nürnberg B, Schwab M, Hofmann U, Haag M. Untargeted stable isotope-resolved metabolomics to assess the effect of PI3Kβ inhibition on metabolic pathway activities in a PTEN null breast cancer cell line. Front Mol Biosci 2022; 9:1004602. [PMID: 36310598 PMCID: PMC9614656 DOI: 10.3389/fmolb.2022.1004602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/28/2022] [Indexed: 11/17/2022] Open
Abstract
The combination of high-resolution LC-MS untargeted metabolomics with stable isotope-resolved tracing is a promising approach for the global exploration of metabolic pathway activities. In our established workflow we combine targeted isotopologue feature extraction with the non-targeted X13CMS routine. Metabolites, detected by X13CMS as differentially labeled between two biological conditions are subsequently integrated into the original targeted library. This strategy enables monitoring of changes in known pathways as well as the discovery of hitherto unknown metabolic alterations. Here, we demonstrate this workflow in a PTEN (phosphatase and tensin homolog) null breast cancer cell line (MDA-MB-468) exploring metabolic pathway activities in the absence and presence of the selective PI3Kβ inhibitor AZD8186. Cells were fed with [U-13C] glucose and treated for 1, 3, 6, and 24 h with 0.5 µM AZD8186 or vehicle, extracted by an optimized sample preparation protocol and analyzed by LC-QTOF-MS. Untargeted differential tracing of labels revealed 286 isotope-enriched features that were significantly altered between control and treatment conditions, of which 19 features could be attributed to known compounds from targeted pathways. Other 11 features were unambiguously identified based on data-dependent MS/MS spectra and reference substances. Notably, only a minority of the significantly altered features (11 and 16, respectively) were identified when preprocessing of the same data set (treatment vs. control in 24 h unlabeled samples) was performed with tools commonly used for label-free (i.e. w/o isotopic tracer) non-targeted metabolomics experiments (Profinder´s batch recursive feature extraction and XCMS). The structurally identified metabolites were integrated into the existing targeted isotopologue feature extraction workflow to enable natural abundance correction, evaluation of assay performance and assessment of drug-induced changes in pathway activities. Label incorporation was highly reproducible for the majority of isotopologues in technical replicates with a RSD below 10%. Furthermore, inter-day repeatability of a second label experiment showed strong correlation (Pearson R2 > 0.99) between tracer incorporation on different days. Finally, we could identify prominent pathway activity alterations upon PI3Kβ inhibition. Besides pathways in central metabolism, known to be changed our workflow revealed additional pathways, like pyrimidine metabolism or hexosamine pathway. All pathways identified represent key metabolic processes associated with cancer metabolism and therapy.
Collapse
Affiliation(s)
- Marcel Lackner
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
| | - Sylvia K. Neef
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
| | - Stefan Winter
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
| | - Sandra Beer-Hammer
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute for Experimental and Clinical Pharmacology and Pharmacogenomics, Interfaculty Center for Pharmacogenomics and Drug Research (ICePhA), University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180), Image-Guided and Functionally Instructed Tumor Therapies, University of Tübingen, Tübingen, Germany
| | - Bernd Nürnberg
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute for Experimental and Clinical Pharmacology and Pharmacogenomics, Interfaculty Center for Pharmacogenomics and Drug Research (ICePhA), University of Tübingen, Tübingen, Germany
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180), Image-Guided and Functionally Instructed Tumor Therapies, University of Tübingen, Tübingen, Germany
- Departments of Clinical Pharmacology and of Pharmacy and Biochemistry, University of Tübingen, Tübingen, Germany
| | - Ute Hofmann
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
| | - Mathias Haag
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
- *Correspondence: Mathias Haag,
| |
Collapse
|
13
|
Introduction of mutant TP53 related genes in metabolic pathways and evaluation their correlation with immune cells, drug resistance and sensitivity. Life Sci 2022; 303:120650. [PMID: 35667517 DOI: 10.1016/j.lfs.2022.120650] [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: 02/12/2022] [Revised: 05/03/2022] [Accepted: 05/16/2022] [Indexed: 01/18/2023]
Abstract
BACKGROUND Although the relationship between TP53 mutation, TP53 metabolism pathways, and tumorigenesis has been investigated, pan-cancer analysis of TP53 mutations and related metabolism pathways is not completely available in common types of human cancers. Thus, this study was going to represent TP53 mutant-related metabolism genes and pathways in a pan-cancer study and investigate the relationship between selected genes and drug resistance. METHODS The DNA-seq data, RNA-seq data, and clinical information of 12 types of cancer were downloaded from the cancer genome atlas (TCGA) database. GSE70479 data were obtained from GEO database for validation of our TCGA data. To evaluate the survival rate of patients, GEPIA2 was applied. The CCLE and GDSC database were used to investigate drug resistance and sensitivity. RESULTS Our findings indicated that TTN, MUC16, and TP53 were present in 12 types of cancer with high level of mutation frequency which abundance of TP53 mutations was higher. Mutant TP53-related (mTP53) pathways and genes including PKM, SLC16A3, HK2, PFKP, PHGDH, and CTSC were obtained from enrichment analysis and interestingly, top pathways were associated with metabolism including glycolysis and mTORC1 pathway. Our results showed the expression of some candidate genes correlated with immune markers, prognosis, and drug resistance. CONCLUSIONS Top mutant genes for 12 cancers were highlighted while TP53 was selected as top mutant gene, and metabolic genes associated with the TP53 mutation were identified that some of which are important in poor prognosis. In doing so, mutations in TP53 could run some metabolic pathways and drug resistance and sensitivity.
Collapse
|
14
|
Kennedy BE, Giacomantonio M, Murphy JP, Cutler S, Sadek M, Konda P, Paulo JA, Pathak GP, Renkens SH, Grieve S, Pol J, Gygi SP, Richardson C, Gaston D, Reiman A, Kroemer G, Elnenaei MO, Gujar SA. NAD+ depletion enhances reovirus-induced oncolysis in multiple myeloma. Mol Ther Oncolytics 2022; 24:695-706. [PMID: 35284625 PMCID: PMC8904403 DOI: 10.1016/j.omto.2022.02.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 02/17/2022] [Indexed: 11/26/2022] Open
Abstract
Cancer cell energy metabolism plays an important role in dictating the efficacy of oncolysis by oncolytic viruses. To understand the role of multiple myeloma metabolism in reovirus oncolysis, we performed semi-targeted mass spectrometry-based metabolomics on 12 multiple myeloma cell lines and revealed a negative correlation between NAD+ levels and susceptibility to oncolysis. Likewise, a negative correlation was observed between the activity of the rate-limiting NAD+ synthesis enzyme NAMPT and oncolysis. Indeed, depletion of NAD+ levels by pharmacological inhibition of NAMPT using FK866 sensitized several myeloma cell lines to reovirus-induced killing. The myelomas that were most sensitive to this combination therapy expressed a functional p53 and had a metabolic and transcriptomic profile favoring mitochondrial metabolism over glycolysis, with the highest synergistic effect in KMS12 cells. Mechanistically, U-13C-labeled glucose flux, extracellular flux analysis, multiplex proteomics, and cell death assays revealed that the reovirus + FK866 combination caused mitochondrial dysfunction and energy depletion, leading to enhanced autophagic cell death in KMS12 cells. Finally, the combination of reovirus and NAD+ depletion achieved greater antitumor effects in KMS12 tumors in vivo and patient-derived CD138+ multiple myeloma cells. These findings identify NAD+ depletion as a potential combinatorial strategy to enhance the efficacy of oncolytic virus-based therapies in multiple myeloma.
Collapse
|
15
|
Dai Z, Liu T, Liu G, Deng Z, Yu P, Wang B, Cen B, Guo L, Zhang J. Identification of Clinical and Tumor Microenvironment Characteristics of Hypoxia-Related Risk Signature in Lung Adenocarcinoma. Front Mol Biosci 2021; 8:757421. [PMID: 34869590 PMCID: PMC8634728 DOI: 10.3389/fmolb.2021.757421] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/22/2021] [Indexed: 12/18/2022] Open
Abstract
Background: Lung cancer is the leading cause of cancer-related death globally. Hypoxia can suppress the activation of the tumor microenvironment (TME), which contributes to distant metastasis. However, the role of hypoxia-mediated TME in predicting the diagnosis and prognosis of lung adenocarcinoma (LUAD) patients remains unclear. Methods: Both RNA and clinical data from the LUAD cohort were downloaded from the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. Both univariate and multivariate Cox regression analyses were used to further screen prognosis-related hypoxia gene clusters. Time-dependent receiver operation characteristic (ROC) curves were established to evaluate the predictive sensitivity and specificity of the hypoxia-related risk signature. The characterization of gene set enrichment analysis (GSEA) and TME immune cell infiltration were further explored to identify hypoxia-related immune infiltration. Results: Eight hypoxia-related genes (LDHA, DCN, PGK1, PFKP, FBP1, LOX, ENO3, and CXCR4) were identified and established to construct a hypoxia-related risk signature. The high-risk group showed a poor overall survival compared to that of the low-risk group in the TCGA and GSE68465 cohorts (p < 0.0001). The AUCs for 1-, 3-, and 5-year overall survival were 0.736 vs. 0.741, 0.656 vs. 0.737, and 0.628 vs. 0.649, respectively. The high-risk group was associated with immunosuppression in the TME. Conclusion: The hypoxia-related risk signature may represent an independent biomarker that can differentiate the characteristics of TME immune cell infiltration and predict the prognosis of LUAD.
Collapse
Affiliation(s)
- Zili Dai
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Diseases, Institute of Respiratory Disease, Guangzhou, China
| | - Taisheng Liu
- Department of Thoracic Surgery, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Guihong Liu
- Department of Radiation Oncology, DongGuan Tungwah Hospital, Dongguan, China
| | - Zhen Deng
- Department of Radiation Oncology, Huizhou Municipal Central Hospital, Huizhou, China
| | - Peng Yu
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Diseases, Institute of Respiratory Disease, Guangzhou, China
| | - Baiyao Wang
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Diseases, Institute of Respiratory Disease, Guangzhou, China
| | - Bohong Cen
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Diseases, Institute of Respiratory Disease, Guangzhou, China
| | - Liyi Guo
- Department of Oncology and Hematology, The Six People's Hospital of Huizhou City, Huiyang Hospital Affiliated to Southern Medical University, Huizhou, China
| | - Jian Zhang
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Diseases, Institute of Respiratory Disease, Guangzhou, China
| |
Collapse
|
16
|
Inaishi T, Shibata M, Ichikawa T, Kanda M, Hayashi M, Soeda I, Takeuchi D, Takano Y, Tsunoda N, Kodera Y, Kikumori T. Platelet isoform of phosphofructokinase accelerates malignant features in breast cancer. Oncol Rep 2021; 47:9. [PMID: 34751415 DOI: 10.3892/or.2021.8220] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 07/06/2021] [Indexed: 11/06/2022] Open
Abstract
The platelet isoform of phosphofructokinase (PFKP) is one of the key enzymes in the glycolytic pathway. PFKP is highly expressed in several cancers, and it has been reported to be involved in the progression of cancer cells. However, its oncological role in breast cancer (BC) remains unclear. The present study aimed to evaluate the function of PFKP in BC cells and its expression level in patients with BC. Firstly, the mRNA and protein expression of PFKP was evaluated in BC and non‑cancerous mammary cell lines. Polymerase chain reaction (PCR) array analysis was conducted to evaluate the correlation between PFKP and 84 cancer‑related genes. Then, PFKP knockdown was conducted using small interfering RNA, and cell proliferation, invasiveness and migration were analyzed. Furthermore, the association between PFKP mRNA expression and clinicopathological factors was investigated in 167 patients with BC. PFKP was highly expressed in estrogen receptor‑negative and human epidermal growth factor receptor 2‑negative BC cell lines. PCR array analysis demonstrated that the expression level of PFKP was significantly correlated with that of transforming growth factor‑β1 and MYC proto‑oncogene. PFKP knockdown significantly decreased the proliferation and invasiveness of MCF7, SK‑BR‑3, and MDA‑MB‑231 cells. Furthermore, cell migration was inhibited in SK‑BR‑3 and MDA‑MB‑231 cells. In the clinical specimens, patients with T2/T3/T4, lymph node metastasis, or stage II/III/IV exhibited higher expression of PFKP mRNA than patients with less severe disease. In conclusion, the present findings indicated that PFKP is involved in promoting tumor‑progressive oncological roles in BC cells across different subtypes and is considered a possible novel therapeutic target for BC.
Collapse
Affiliation(s)
- Takahiro Inaishi
- Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine, Nagoya 466‑8550, Japan
| | - Masahiro Shibata
- Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine, Nagoya 466‑8550, Japan
| | - Takahiro Ichikawa
- Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine, Nagoya 466‑8550, Japan
| | - Mitsuro Kanda
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya 466‑8550, Japan
| | - Masamichi Hayashi
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya 466‑8550, Japan
| | - Ikumi Soeda
- Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine, Nagoya 466‑8550, Japan
| | - Dai Takeuchi
- Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine, Nagoya 466‑8550, Japan
| | - Yuko Takano
- Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine, Nagoya 466‑8550, Japan
| | - Nobuyuki Tsunoda
- Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine, Nagoya 466‑8550, Japan
| | - Yasuhiro Kodera
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya 466‑8550, Japan
| | - Toyone Kikumori
- Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine, Nagoya 466‑8550, Japan
| |
Collapse
|
17
|
Liu B, Li F, Liu M, Xu Z, Gao B, Wang Y, Zhou H. Prognostic Roles of Phosphofructokinase Platelet in Clear Cell Renal Cell Carcinoma and Correlation with Immune Infiltration. Int J Gen Med 2021; 14:3645-3658. [PMID: 34321910 PMCID: PMC8312753 DOI: 10.2147/ijgm.s321337] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 07/08/2021] [Indexed: 12/24/2022] Open
Abstract
Background Abnormal expression of phosphofructokinase platelet (PFKP) has been reported in various cancer types. However, the role of PFKP in clear cell renal cell carcinoma (ccRCC) remains unclear. Methods In this study, the PFKP expression levels in various cancers were systemically described by integrating multiple kinds of publicly available databases. The relationship between PFKP expression and clinical prognosis of ccRCC patients was analyzed based on the TCGA database. Furthermore, PFKP-related genes and the top 10 hub genes were identified. The enrichment analysis, PPI network, and the relationship between PFKP and tumor-infiltrating immune cells were conducted to explore why PFKP was associated with clinical outcomes in ccRCC patients. Results PFKP was significantly highly expressed in kidney cancer, especially in ccRCC. Moreover, patients with low expression of PFKP were correlated with poor 5-year and 10-year overall survival (OS) (P < 0.05). Low PFKP expression was a risk factor associated with decreased OS in subgroups including males, females, grade 3–4, and stage III–IV (all P < 0.05). GO and KEGG enrichment analyses showed that 10 hub genes were mainly enriched in the tumor immune response. Finally, PFKP expression level was highly correlated with the infiltration of B cell, CD8+ T cell, CD4+ T cell, macrophage, neutrophil, and dendritic cell. Conclusion In short, our findings suggested that PFKP is highly expressed in ccRCC significantly and facilitated tumor immune response which in turn associated with a good prognosis.
Collapse
Affiliation(s)
- Bin Liu
- Department of Urology, the First Hospital of Jilin University, Changchun, 130021, Jilin, People's Republic of China
| | - Faping Li
- Department of Urology, the First Hospital of Jilin University, Changchun, 130021, Jilin, People's Republic of China
| | - Mingdi Liu
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, Jilin, People's Republic of China
| | - Zhixiang Xu
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, Jilin, People's Republic of China
| | - Baoshan Gao
- Department of Urology, the First Hospital of Jilin University, Changchun, 130021, Jilin, People's Republic of China
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, Jilin, People's Republic of China
| | - Honglan Zhou
- Department of Urology, the First Hospital of Jilin University, Changchun, 130021, Jilin, People's Republic of China
| |
Collapse
|
18
|
Loss of the transcriptional repressor Rev-erbα upregulates metabolism and proliferation in cultured mouse embryonic fibroblasts. Sci Rep 2021; 11:12356. [PMID: 34117285 PMCID: PMC8196003 DOI: 10.1038/s41598-021-91516-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 05/19/2021] [Indexed: 12/04/2022] Open
Abstract
The transcriptional repressor Rev-erbα is known to down-regulate fatty acid metabolism and gluconeogenesis gene expression. In animal models, disruption of Rev-erbα results in global changes in exercise performance, oxidative capacity, and blood glucose levels. However, the complete extent to which Rev-erbα-mediated transcriptional repression of metabolism impacts cell function remains unknown. We hypothesized that loss of Rev-erbα in a mouse embryonic fibroblast (MEF) model would result in global changes in metabolism. MEFs lacking Rev-erbα exhibited a hypermetabolic phenotype, demonstrating increased levels of glycolysis and oxidative phosphorylation. Rev-erbα deletion increased expression of hexokinase II, transketolase, and ribose-5-phosphate isomerase genes involved in glycolysis and the pentose phosphate pathway (PPP), and these effects were not mediated by the transcriptional activator BMAL1. Upregulation of oxidative phosphorylation was not accompanied by an increase in mitochondrial biogenesis or numbers. Rev-erbα repressed proliferation via glycolysis, but not the PPP. When treated with H2O2, cell viability was reduced in Rev-erbα knockout MEFs, accompanied by increased ratio of oxidized/reduced NADPH, suggesting that perturbation of the PPP reduces capacity to mount an antioxidant defense. These findings uncover novel mechanisms by which glycolysis and the PPP are modulated through Rev-erbα, and provide new insights into how Rev-erbα impacts proliferation.
Collapse
|
19
|
Vigil-Garcia M, Demkes CJ, Eding JEC, Versteeg D, de Ruiter H, Perini I, Kooijman L, Gladka MM, Asselbergs FW, Vink A, Harakalova M, Bossu A, van Veen TAB, Boogerd CJ, van Rooij E. Gene expression profiling of hypertrophic cardiomyocytes identifies new players in pathological remodelling. Cardiovasc Res 2021; 117:1532-1545. [PMID: 32717063 PMCID: PMC8152696 DOI: 10.1093/cvr/cvaa233] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 05/15/2020] [Accepted: 07/22/2020] [Indexed: 01/30/2023] Open
Abstract
AIMS Pathological cardiac remodelling is characterized by cardiomyocyte (CM) hypertrophy and fibroblast activation, which can ultimately lead to maladaptive hypertrophy and heart failure (HF). Genome-wide expression analysis on heart tissue has been instrumental for the identification of molecular mechanisms at play. However, these data were based on signals derived from all cardiac cell types. Here, we aimed for a more detailed view on molecular changes driving maladaptive CM hypertrophy to aid in the development of therapies to reverse pathological remodelling. METHODS AND RESULTS Utilizing CM-specific reporter mice exposed to pressure overload by transverse aortic banding and CM isolation by flow cytometry, we obtained gene expression profiles of hypertrophic CMs in the more immediate phase after stress, and CMs showing pathological hypertrophy. We identified subsets of genes differentially regulated and specific for either stage. Among the genes specifically up-regulated in the CMs during the maladaptive phase we found known stress markers, such as Nppb and Myh7, but additionally identified a set of genes with unknown roles in pathological hypertrophy, including the platelet isoform of phosphofructokinase (PFKP). Norepinephrine-angiotensin II treatment of cultured human CMs induced the secretion of N-terminal-pro-B-type natriuretic peptide (NT-pro-BNP) and recapitulated the up-regulation of these genes, indicating conservation of the up-regulation in failing CMs. Moreover, several genes induced during pathological hypertrophy were also found to be increased in human HF, with their expression positively correlating to the known stress markers NPPB and MYH7. Mechanistically, suppression of Pfkp in primary CMs attenuated stress-induced gene expression and hypertrophy, indicating that Pfkp is an important novel player in pathological remodelling of CMs. CONCLUSION Using CM-specific transcriptomic analysis, we identified novel genes induced during pathological hypertrophy that are relevant for human HF, and we show that PFKP is a conserved failure-induced gene that can modulate the CM stress response.
Collapse
MESH Headings
- Animals
- Cardiac Myosins/genetics
- Cardiac Myosins/metabolism
- Cardiomegaly/genetics
- Cardiomegaly/metabolism
- Cardiomegaly/pathology
- Cardiomegaly/physiopathology
- Cells, Cultured
- Disease Models, Animal
- Fibrosis
- Gene Expression Profiling
- Gene Expression Regulation
- Humans
- Male
- Mice, Inbred C57BL
- Mice, Transgenic
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Myosin Heavy Chains/genetics
- Myosin Heavy Chains/metabolism
- Natriuretic Peptide, Brain/genetics
- Natriuretic Peptide, Brain/metabolism
- Phosphofructokinase-1, Type C/genetics
- Phosphofructokinase-1, Type C/metabolism
- Transcriptome
- Ventricular Remodeling/genetics
- Mice
Collapse
Affiliation(s)
- Marta Vigil-Garcia
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands
| | - Charlotte J Demkes
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands
- Department of Cardiology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Joep E C Eding
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands
| | - Danielle Versteeg
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands
| | - Hesther de Ruiter
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands
| | - Ilaria Perini
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands
| | - Lieneke Kooijman
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands
| | - Monika M Gladka
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands
| | - Folkert W Asselbergs
- Department of Cardiology, University Medical Centre Utrecht, Utrecht, The Netherlands
- Institute of Cardiovascular Science, Faculty of Population Health Sciences, University College London, London, UK
- Health Data Research UK and Institute of Health Informatics, University College London, London, UK
| | - Aryan Vink
- Department of Pathology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Magdalena Harakalova
- Department of Cardiology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Alexander Bossu
- Department of Medical Physiology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Toon A B van Veen
- Department of Medical Physiology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Cornelis J Boogerd
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands
| | - Eva van Rooij
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands
- Department of Cardiology, University Medical Centre Utrecht, Utrecht, The Netherlands
| |
Collapse
|
20
|
Udayaraja GK, Arnold Emerson I. Network-based gene deletion analysis identifies candidate genes and molecular mechanism involved in clear cell renal cell carcinoma. J Genet 2021. [DOI: 10.1007/s12041-021-01260-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
21
|
Platelet isoform of phosphofructokinase promotes aerobic glycolysis and the progression of non‑small cell lung cancer. Mol Med Rep 2020; 23:74. [PMID: 33236133 PMCID: PMC7716410 DOI: 10.3892/mmr.2020.11712] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 10/19/2020] [Indexed: 12/13/2022] Open
Abstract
The platelet isoform of phosphofructokinase (PFKP) is a rate-limiting enzyme involved in glycolysis that serves an important role in various types of cancer. The aim of the present study was to explore the specific regulatory relationship between PFKP and non-small cell lung cancer (NSCLC) progression. PFKP expression in NSCLC tissues and corresponding adjacent tissues was detected using reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and immunohistochemical analysis. PFKP expression in human bronchial epithelial cells (16HBE) and NSCLC cells (H1299, H23 and A549) was also detected using RT-qPCR. Cell proliferation was detected by Cell Counting Kit-8 and colony formation assays. Transwell invasion and wound healing assays, and flow cytometry were used to detect cell invasion, migration and apoptosis, respectively. The expression levels of glycolysis-associated enzymes (hexokinase-2, lactate dehydrogenase A and glucose transporter-1), epithelial-mesenchymal transition-related proteins (N-cadherin, vimentin and E-cadherin) and apoptosis-related proteins (caspase-3 and B-cell lymphoma-2) were detected by western blotting. Glucose uptake, lactate production and the adenosine trisphosphate/adenosine diphosphate ratio were measured using the corresponding kits. The results of the present study demonstrated that PFKP expression was upregulated in NSCLC tissues and cells, and PFKP expression was related to lymph node metastasis and histological grade. In addition, overexpression of PFKP inhibited cell apoptosis, and promoted proliferation, migration, invasion and glycolysis of H1299 cells, whereas knockdown of PFKP had the opposite effects. In conclusion, PFKP inhibited cell apoptosis, and promoted proliferation, migration, invasion and glycolysis of NSCLC cells; these findings may lay the foundation for novel treatments of NSCLC.
Collapse
|
22
|
Keuls RA, Kojima K, Lozzi B, Steele JW, Chen Q, Gross SS, Finnell RH, Parchem RJ. MiR-302 Regulates Glycolysis to Control Cell-Cycle during Neural Tube Closure. Int J Mol Sci 2020; 21:E7534. [PMID: 33066028 PMCID: PMC7589003 DOI: 10.3390/ijms21207534] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 09/26/2020] [Accepted: 10/06/2020] [Indexed: 01/03/2023] Open
Abstract
Neural tube closure is a critical early step in central nervous system development that requires precise control of metabolism to ensure proper cellular proliferation and differentiation. Dysregulation of glucose metabolism during pregnancy has been associated with neural tube closure defects (NTDs) in humans suggesting that the developing neuroepithelium is particularly sensitive to metabolic changes. However, it remains unclear how metabolic pathways are regulated during neurulation. Here, we used single-cell mRNA-sequencing to analyze expression of genes involved in metabolism of carbon, fats, vitamins, and antioxidants during neurulation in mice and identify a coupling of glycolysis and cellular proliferation to ensure proper neural tube closure. Using loss of miR-302 as a genetic model of cranial NTD, we identify misregulated metabolic pathways and find a significant upregulation of glycolysis genes in embryos with NTD. These findings were validated using mass spectrometry-based metabolite profiling, which identified increased glycolytic and decreased lipid metabolites, consistent with a rewiring of central carbon traffic following loss of miR-302. Predicted miR-302 targets Pfkp, Pfkfb3, and Hk1 are significantly upregulated upon NTD resulting in increased glycolytic flux, a shortened cell cycle, and increased proliferation. Our findings establish a critical role for miR-302 in coordinating the metabolic landscape of neural tube closure.
Collapse
Affiliation(s)
- Rachel A. Keuls
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA;
- Center for Cell and Gene Therapy, Stem Cells and Regenerative Medicine Center, One Baylor Plaza, Houston, TX 77030, USA;
| | - Karin Kojima
- Center for Cell and Gene Therapy, Stem Cells and Regenerative Medicine Center, One Baylor Plaza, Houston, TX 77030, USA;
| | - Brittney Lozzi
- Genetics and Genomics Graduate Program, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA;
| | - John W. Steele
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; (J.W.S.); (R.H.F.)
- Center for Precision Environmental Health, Department of Molecular and Cellular Biology and Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Qiuying Chen
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA; (Q.C.); (S.S.G.)
| | - Steven S. Gross
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA; (Q.C.); (S.S.G.)
| | - Richard H. Finnell
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; (J.W.S.); (R.H.F.)
- Center for Precision Environmental Health, Department of Molecular and Cellular Biology and Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ronald J. Parchem
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA;
- Center for Cell and Gene Therapy, Stem Cells and Regenerative Medicine Center, One Baylor Plaza, Houston, TX 77030, USA;
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; (J.W.S.); (R.H.F.)
| |
Collapse
|
23
|
Sánchez-Calabuig MJ, Fernández-González R, Hamdi M, Smits K, López-Cardona AP, Serres C, Macías-García B, Gutiérrez-Adán A. A high glucose concentration during early stages of in vitro equine embryo development alters expression of genes involved in glucose metabolism. Equine Vet J 2020; 53:787-795. [PMID: 32881040 DOI: 10.1111/evj.13342] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 07/17/2020] [Accepted: 08/27/2020] [Indexed: 01/30/2023]
Abstract
BACKGROUND Equine embryos exhibit an unusual pattern of glucose tolerance in vitro and are currently cultured in hyperglycaemic conditions. OBJECTIVE Our main objective was to analyse the effect of different glucose concentrations on in vitro-produced equine embryo development and quality. STUDY DESIGN Experiments comparing in vitro and in vivo produced embryos. METHODS Oocytes (n = 641) were collected from post-mortem ovaries, matured in vitro and fertilised by intracytoplasmic sperm injection (ICSI). Embryo culture was divided from Day 0 to Day 4 and from Day 4 to Day 9 in three groups: 5-10 (5 and 10 mmol/L glucose respectively; n = 87); 5-17 (5 and 17.5 mmol/L; n = 66); and 10-17 (10 and 17.5 mmol/L; n = 117). A control group of 20 in vivo produced blastocysts was included. Cleavage and blastocyst rates were evaluated and embryos were snap-frozen for analysis of the relative mRNA expression of genes related to mitochondrial function, DNA methylation, apoptosis, glucose transport and metabolism. RESULTS No differences were observed in the cleavage or blastocyst rates among in vitro groups. Under high glucose conditions in vitro (10-17 group), BAX/BCL2 was higher, and PFKP, LDHA and COX2 were overexpressed compared to all other groups. The two groups with 5 mmol/L glucose concentration during the first culture stage (5-10 and 5-17) displayed similar patterns which differed to the 10-17 group. MAIN LIMITATIONS Conclusions related to embryo quality are based on gene expression patterns. Transfer of in vitro-produced embryos would reveal whether the observed differences improve embryo developmental competence. CONCLUSIONS Five mM glucose during the first days of culture seems to be preferable to avoid over-activation of embryonic glycolytic pathways. Further studies are necessary to determine whether this improves embryo developmental competence.
Collapse
Affiliation(s)
- María J Sánchez-Calabuig
- Department of Animal Medicine and Surgery, Faculty of Veterinary Science, University Complutense of Madrid, Madrid, Spain.,Department of Animal Reproduction, INIA, Madrid, Spain
| | | | - Meriem Hamdi
- Department of Animal Reproduction, INIA, Madrid, Spain
| | - Katrien Smits
- Department of Obstetrics, Reproduction and Herd Health, Ghent University, Ghent, Belgium
| | - Angela P López-Cardona
- Department of Animal Reproduction, INIA, Madrid, Spain.,Grupo de Investigación (GI) - Biogénesis, Universidad de Antioquia, Medellín, Colombia
| | - Consuelo Serres
- Department of Animal Medicine and Surgery, Faculty of Veterinary Science, University Complutense of Madrid, Madrid, Spain
| | - Beatriz Macías-García
- Animal Medicine Department, Faculty of Veterinary Sciences, University of Extremadura, Cáceres, Spain.,Research Group of Intracellular Signalling and Technology of Reproduction (SINTREP), Research Institute of Biotechnology in Livestock and Cynegetic (INBIO G+C), University of Extremadura, Cáceres, Spain
| | | |
Collapse
|
24
|
Qin Y, Zheng B, Yang GS, Yang HJ, Zhou J, Yang Z, Zhang XH, Zhao HY, Shi JH, Wen JK. Salvia miltiorrhiza-Derived Sal-miR-58 Induces Autophagy and Attenuates Inflammation in Vascular Smooth Muscle Cells. MOLECULAR THERAPY-NUCLEIC ACIDS 2020; 21:492-511. [PMID: 32679544 PMCID: PMC7360890 DOI: 10.1016/j.omtn.2020.06.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/15/2020] [Accepted: 06/18/2020] [Indexed: 12/15/2022]
Abstract
Autophagy is associated with the cytoprotection of physiological processes against inflammation and oxidative stress. Salvia miltiorrhiza possesses cardiovascular protective actions and has powerful anti-oxidative and anti-inflammatory effects; however, whether and how Salvia miltiorrhiza-derived microRNAs (miRNAs) protect vascular smooth muscle cells (VSMCs) by inducing autophagy across species are unknown. We first screened and identified Sal-miR-58 from Salvia miltiorrhiza as a natural autophagy inducer. Synthetic Sal-miR-58 suppresses chronic angiotensin II (Ang II) infusion-induced abdominal aortic aneurysm (AAA) formation in mice, as well as induces autophagy in VSMCs and attenuates the inflammatory response elicited by Ang II in vivo and in vitro. Mechanistically, Sal-miR-58 downregulates Krüppel-like factor 3 (KLF3) expression through direct binding to the 3' UTR of KLF3, which in turn relieves KLF3 repression of E3 ubiquitin ligase neural precursor cell-expressed developmentally downregulated 4-like (NEDD4L) expression, whereas NEDD4L upregulation increases the ubiquitination and degradation of the platelet isoform of phosphofructokinase (PFKP), subsequently leading to a decrease in the activation of Akt/mammalian target of rapamycin (mTOR) signaling and facilitating VSMC autophagy induced by Sal-miR-58 in the context of chronic Ang II stimulation and aneurysm formation. Our results provide the first evidence that plant-derived Sal-miR-58 induces autophagy and attenuates inflammation in VSMCs through cross-species modulation of the KLF3/NEDD4L/PFKP regulatory pathway.
Collapse
Affiliation(s)
- Yan Qin
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, Shijiazhuang 050017, China; Department of Central Laboratory, Affiliated Hospital of Hebei University, Baoding 071000, China; Department of Life Science and Green Development, Hebei University, Baoding 071000, China
| | - Bin Zheng
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, Shijiazhuang 050017, China
| | - Gao-Shan Yang
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, Shijiazhuang 050017, China; Department of Biochemistry and Molecular Biology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Hao-Jie Yang
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, Shijiazhuang 050017, China
| | - Jing Zhou
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, Shijiazhuang 050017, China; Department of Endocrine, The Second Hospital of Hebei Medical University, Shijiazhuang 050005, China
| | - Zhan Yang
- Department of Science and Technology, The Second Hospital of Hebei Medical University, Shijiazhuang 050005, China
| | - Xin-Hua Zhang
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, Shijiazhuang 050017, China
| | - Hong-Ye Zhao
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, Shijiazhuang 050017, China
| | - Jian-Hong Shi
- Department of Central Laboratory, Affiliated Hospital of Hebei University, Baoding 071000, China; Department of Life Science and Green Development, Hebei University, Baoding 071000, China
| | - Jin-Kun Wen
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, Shijiazhuang 050017, China.
| |
Collapse
|
25
|
PFKP is transcriptionally repressed by BRCA1/ZBRK1 and predicts prognosis in breast cancer. PLoS One 2020; 15:e0233750. [PMID: 32470015 PMCID: PMC7259711 DOI: 10.1371/journal.pone.0233750] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 05/12/2020] [Indexed: 12/24/2022] Open
Abstract
Objectives The present study aims to elucidate the underlying mechanism how PFKP is regulated by BRCA1 and the clinical significance of PFKP in breast cancer. Methods MEF-BRCA1△/△ and the wild type counterpart MEF-BRCA1+/+ cell lines were used to test the sensitivity of glucose depletion in culture medium. Glucose Assay Kit was used to quantify glucose levels in cultural supernatant and cell lysate. Real time PCR was used to measure the mRNA expression levels of genes. Western blot was used to detect protein levels. Chromatin immunoprecipitation was used to verify the bindings between transcription factors and DNA elements. Luciferase reporter assay was performed to determine the transcriptional activity. Histochemistry assay was performed on tissue microarray. Results We found that MEF-BRCA1△/△ cells consumed more glucose and were more vulnerable to glucose-deprived culture medium. The mRNA profiles and qPCR assay of MEF-BRCA1△/△ and MEF-BRCA1+/+ cells revealed that PFKP, the rate-limiting enzyme of glycolysis, was significantly upregulated in MEF-BRCA1△/△ cells. Consistently, the repressive effects of BRCA1 on PFKP were confirmed by overexpression or knockdown of BRCA1. Moreover, we also demonstrated that PFKP was suppressed by ZBRK1 as well, which was the co-repression partner of BRCA1. Mechanistically, we figured out that BRCA1 formed a transcriptional repression complex with ZBRK1 on the promoter of PFKP and consequently restrained its expression. Importantly, the expression levels of PFKP were demonstrated to associate with poor survival of patients with breast cancer. Conclusion Our study provided a new insight into the dysregulation of glycolysis in breast cancer, which might be partially due to the deficiency of BRCA1/ZBRK1 axis and subsequently reversed the transcriptional repressive effect on PFKP. We also found that PFKP overexpressed in a subset of breast cancer patients and could serve as a prognostic factor, which represented a potential target for BC therapy.
Collapse
|
26
|
Wang Z, Sun X, Gao L, Guo X, Feng C, Lian W, Deng K, Xing B. Comprehensive identification of a two-genesignature as a novel potential prognostic model for patients with medulloblastoma. Am J Transl Res 2020; 12:1600-1613. [PMID: 32509164 PMCID: PMC7270006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 05/08/2020] [Indexed: 06/11/2023]
Abstract
Medulloblastoma is one of the most common malignant pediatric brain tumors and has a poor prognosis and high mortality. We investigated the prognostic significance of specific gene signatures and established a novel prognostic model for medulloblastoma patients. Ninety-seven differentially expressed genes between 69 medulloblastoma samples and 4 normal cerebellum samples were identified using the GSE68956 dataset. Univariate and multivariate Cox regression analyses revealed optimal prognosis-related genes, of which PFKP and STXBP1 exhibited significant prognostic values. A risk score model was then established to assess the prognostic value of the gene signature. Kaplan-Meier survival analysis demonstrated that patients with a high risk score had significantly poorer overall survival (OS, log-rank P = 0.003308). The concordance index (C-index) of the two-gene prognostic model for OS prediction was 0.752 (95% CI, 0.740-0.764). The area under the receiver operating characteristic curve (AUC) values for predicting 3-year and 5-year survival were 0.726 and 0.730, respectively. The risk score model was further validated in the ICGC cohort and PUMCH cohort using quantitative real-time polymerase chain reaction (qRT-PCR). Cox regression analyses were performed to assess the two-gene risk score model, metastasis stage, and chemotherapy as independent prognostic factors for medulloblastoma. The C-index of the comprehensive prognostic model composed of the two-gene signature integrated with clinicopathological features for predicting OS was 0.823 (95% CI, 0.739-0.907). The AUCs of the comprehensive prognostic model for predicting 3-year and 5-year survival were 0.774 and 0.759, respectively. Thus, the two-gene risk score model is a promising prognostic biomarker for medulloblastoma.
Collapse
Affiliation(s)
- Zihao Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeDongcheng District, Beijing, P. R. China
| | - Xuesong Sun
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and TherapyGuangzhou, P. R. China
- Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer CenterGuangzhou, P. R. China
| | - Lu Gao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeDongcheng District, Beijing, P. R. China
| | - Xiaopeng Guo
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeDongcheng District, Beijing, P. R. China
| | - Chenzhe Feng
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeDongcheng District, Beijing, P. R. China
| | - Wei Lian
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeDongcheng District, Beijing, P. R. China
| | - Kan Deng
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeDongcheng District, Beijing, P. R. China
| | - Bing Xing
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeDongcheng District, Beijing, P. R. China
| |
Collapse
|
27
|
PFKP is highly expressed in lung cancer and regulates glucose metabolism. Cell Oncol (Dordr) 2020; 43:617-629. [PMID: 32219704 DOI: 10.1007/s13402-020-00508-6] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2020] [Indexed: 12/23/2022] Open
Abstract
PURPOSE Although it has been reported that up-regulation of phosphofructokinase (PFK) expression is a major feature of malignant tumors, the role of platelet type PFK (PFKP) in tumor initiation and progression is as yet poorly understood. The objective of this study was to evaluate PFKP expression in lung cancer and its effect on glycolysis, and to explore correlations between PFKP expression levels and clinical lung cancer patient features. METHODS PFKP mRNA expression levels in cancer tissues and adjacent normal tissues were compared using The Cancer Genome Atlas (TCGA) database. PFKP mRNA and protein expression levels in fresh lung cancer tissues and cell lines were assessed using quantitative real-time PCR and Western blotting. Immunohistochemistry (IHC) was used to assess PFKP expression in 150 archival lung adenocarcinoma samples, after which follow-up data and their correlations with clinical features and patient prognosis were evaluated. A retroviral shRNA-mediated method was used to construct stable cell lines expressing low levels of PFKP. Glucose, lactate and adenosine triphosphate concentrations in the cell culture supernatants were determined using enzymatic, spectrophotometric and enzyme-linked immunosorbent (ELISA) assays, respectively. The effect of PFKP expression on the proliferation of lung cancer cells was assessed using colony forming, MTT and flow cytometry assays, respectively. Finally, data from tissue samples of 533 patients with lung adenocarcinoma and 502 patients with lung squamous cell carcinoma were downloaded from the TCGA database, after which pathway and gene correlation information was retrieved using gene set enrichment analyses. RESULTS We found that PFKP was highly expressed in lung cancer tissues and cell lines. Using IHC we found that PFKP was highly expressed in primary lung adenocarcinoma tissues and that a high expression was associated with a poor prognosis. Clinical data analysis revealed that the PFKP expression levels correlated with tumor size and patient survival. Lung cancer cell lines with decreased PFKP expression levels showed significant decreases in glucose uptake rates, lactate levels and adenosine triphosphate concentrations. They also exhibited significantly decreased proliferation rates, colony forming abilities and increased G2/M cell cycle phase percentages. Gene set enrichment analysis revealed that multiple pathways, including glycolytic pathways, may be involved in the regulation PFKP. CONCLUSIONS Our data indicate that PFKP is highly expressed in lung cancer tissues and cell lines and is associated with tumor size and patient prognosis. As such, PFKP may serve as a prognostic biomarker. We also found that PFKP regulates the level of glycolysis in lung cancer cells and is associated with lung cancer cell proliferation. These data may be instrumental for the design of new lung cancer treatment options.
Collapse
|
28
|
Effective electrochemotherapy with curcumin in MDA-MB-231-human, triple negative breast cancer cells: A global proteomics study. Bioelectrochemistry 2020; 131:107350. [DOI: 10.1016/j.bioelechem.2019.107350] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 08/19/2019] [Accepted: 08/19/2019] [Indexed: 11/22/2022]
|
29
|
Zhang X, Li J, Ghoshal K, Fernandez S, Li L. Identification of a Subtype of Hepatocellular Carcinoma with Poor Prognosis Based on Expression of Genes within the Glucose Metabolic Pathway. Cancers (Basel) 2019; 11:E2023. [PMID: 31847435 PMCID: PMC6966574 DOI: 10.3390/cancers11122023] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/11/2019] [Accepted: 12/11/2019] [Indexed: 12/13/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most prevalent primary cancer and a highly aggressive liver malignancy. Liver cancer cells reprogram their metabolism to meet their needs for rapid proliferation and tumor growth. In the present study, we investigated the alterations in the expression of the genes involved in glucose metabolic pathways as well as their association with the clinical stage and survival of HCC patients. We found that the expressions of around 30% of genes involved in the glucose metabolic pathway are consistently dysregulated with a predominant down-regulation in HCC tumors. Moreover, the differentially expressed genes are associated with an advanced clinical stage and a poor prognosis. More importantly, unsupervised clustering analysis with the differentially expressed genes that were also associated with overall survival (OS) revealed a subgroup of patients with a worse prognosis including reduced OS, disease specific survival, and recurrence-free survival. This aggressive subtype had significantly increased expression of stemness-related genes and down-regulated metabolic genes, as well as increased immune infiltrates that contribute to a poor prognosis. Collectively, this integrative study indicates that expressions of the glucose metabolic genes could be used as potential prognostic markers and/or therapeutic targets, which might be helpful in developing precise treatment for patients with HCC.
Collapse
Affiliation(s)
- Xiaoli Zhang
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43201, USA; (J.L.); (S.F.); (L.L.)
| | - Jin Li
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43201, USA; (J.L.); (S.F.); (L.L.)
| | - Kalpana Ghoshal
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH 43201, USA;
| | - Soledad Fernandez
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43201, USA; (J.L.); (S.F.); (L.L.)
| | - Lang Li
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43201, USA; (J.L.); (S.F.); (L.L.)
| |
Collapse
|
30
|
Fujiwara H, Tateishi K, Misumi K, Hayashi A, Igarashi K, Kato H, Nakatsuka T, Suzuki N, Yamamoto K, Kudo Y, Hayakawa Y, Nakagawa H, Tanaka Y, Ijichi H, Kogure H, Nakai Y, Isayama H, Hasegawa K, Fukayama M, Soga T, Koike K. Mutant IDH1 confers resistance to energy stress in normal biliary cells through PFKP-induced aerobic glycolysis and AMPK activation. Sci Rep 2019; 9:18859. [PMID: 31827136 PMCID: PMC6906335 DOI: 10.1038/s41598-019-55211-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Accepted: 11/22/2019] [Indexed: 02/07/2023] Open
Abstract
Metabolism is a critical regulator of cell fate determination. Recently, the significance of metabolic reprogramming in environmental adaptation during tumorigenesis has attracted much attention in cancer research. Recurrent mutations in the isocitrate dehydrogenase (IDH) 1 or 2 genes have been identified in several cancers, including intrahepatic cholangiocarcinoma (ICC). Mutant IDHs convert α-ketoglutarate (α-KG) to 2-hydroxyglutarate (2-HG), which affects the activity of multiple α-KG-dependent dioxygenases including histone lysine demethylases. Although mutant IDH can be detected even in the early stages of neoplasia, how IDH mutations function as oncogenic drivers remains unclear. In this study, we aimed to address the biological effects of IDH1 mutation using intrahepatic biliary organoids (IBOs). We demonstrated that mutant IDH1 increased the formation of IBOs as well as accelerated glucose metabolism. Gene expression analysis and ChIP results revealed the upregulation of platelet isoform of phosphofructokinase-1 (PFKP), which is a rate-limiting glycolytic enzyme, through the alteration of histone modification. Knockdown of the Pfkp gene alleviated the mutant IDH1-induced increase in IBO formation. Notably, the high expression of PFKP was observed more frequently in patients with IDH-mutant ICC compared to in those with wild-type IDH (p < 0.01, 80.9% vs. 42.5%, respectively). Furthermore, IBOs expressing mutant IDH1 survived the suppression of ATP production caused by growth factor depletion and matrix detachment by retaining high ATP levels through 5' adenosine monophosphate-activated protein kinase (AMPK) activation. Our findings provide a systematic understanding as to how mutant IDH induces tumorigenic preconditioning by metabolic rewiring in intrahepatic cholangiocytes.
Collapse
Affiliation(s)
- Hiroaki Fujiwara
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
- Division of Gastroenterology, The Institute for Adult Diseases, Asahi Life Foundation, 2-2-6 Bakurocho, Chuo-ku, Tokyo, 103-0002, Japan
| | - Keisuke Tateishi
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Kento Misumi
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Akimasa Hayashi
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Kaori Igarashi
- Institute for Advanced Biosciences, Keio University, 246-2 Kakuganji, Tsuruoka, Yamagata, 997-0052, Japan
| | - Hiroyuki Kato
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Takuma Nakatsuka
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Nobumi Suzuki
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Keisuke Yamamoto
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yotaro Kudo
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yoku Hayakawa
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Hayato Nakagawa
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yasuo Tanaka
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Hideaki Ijichi
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Hirofumi Kogure
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yosuke Nakai
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Hiroyuki Isayama
- Department of Gastroenterology, Graduate School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Kiyoshi Hasegawa
- Hepato-Biliary-Pancreatic Division, Department of Surgery, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Masashi Fukayama
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, 246-2 Kakuganji, Tsuruoka, Yamagata, 997-0052, Japan
| | - Kazuhiko Koike
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| |
Collapse
|
31
|
Zhang P, Fu H, Du S, Wang F, Yang J, Cai W, Liu D. Click RNA for Rapid Capture and Identification of Intracellular MicroRNA Targets. Anal Chem 2019; 91:15740-15747. [PMID: 31714070 DOI: 10.1021/acs.analchem.9b03943] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Rapid capture and identification of the intracellular target genes of microRNAs (miRNAs) are the key to understanding miRNA functions and development of RNA-based therapeutics. However, developing biochemical tools that can fish out the target genes of miRNAs in live cells is a significant technical challenge. Here, we report a remarkably simple yet powerful technology capable of loading virtually any miRNA into Ago2 of the RNA-induced silencing complexes (RISCs). This surprising discovery enables rapid capture and identification of target mRNAs and long noncoding RNAs. It is achieved by linking dibenzocyclooctyne (DBCO), a classical chemical moiety in copper-free click chemistry, to the 3' end of miRNAs. DBCO serves as a high-affinity tag to the Ago2 protein, thus boosting the formation of RISCs with miRNA target genes in living cells. Upon cell lysing, DBCO's routine function in click chemistry allows rapid enrichment of target genes for analysis without the need of additional molecular handles. A series of miR-21 and miR-27a target genes that were previously unknown were pulled down from various cell lines and identified with qRT-PCR, demonstrating the utility of this innovative technology in both transcriptomic research and RNA-based studies.
Collapse
Affiliation(s)
- Pengjuan Zhang
- College of Chemistry, Research Center for Analytical Sciences, State Key Laboratory of Medicinal Chemical Biology, and Tianjin Key Laboratory of Molecular Recognition and Biosensing , Nankai University , Tianjin 300071 , China.,Hebei University of Environmental Engineering , Qinhuangdao 066102 , Hebei , China
| | - Haohao Fu
- College of Chemistry, Research Center for Analytical Sciences, State Key Laboratory of Medicinal Chemical Biology, and Tianjin Key Laboratory of Molecular Recognition and Biosensing , Nankai University , Tianjin 300071 , China
| | - Shuangli Du
- College of Chemistry, Research Center for Analytical Sciences, State Key Laboratory of Medicinal Chemical Biology, and Tianjin Key Laboratory of Molecular Recognition and Biosensing , Nankai University , Tianjin 300071 , China
| | - Fengchao Wang
- College of Chemistry, Research Center for Analytical Sciences, State Key Laboratory of Medicinal Chemical Biology, and Tianjin Key Laboratory of Molecular Recognition and Biosensing , Nankai University , Tianjin 300071 , China
| | - Jie Yang
- College of Chemistry, Research Center for Analytical Sciences, State Key Laboratory of Medicinal Chemical Biology, and Tianjin Key Laboratory of Molecular Recognition and Biosensing , Nankai University , Tianjin 300071 , China
| | - Wensheng Cai
- College of Chemistry, Research Center for Analytical Sciences, State Key Laboratory of Medicinal Chemical Biology, and Tianjin Key Laboratory of Molecular Recognition and Biosensing , Nankai University , Tianjin 300071 , China
| | - Dingbin Liu
- College of Chemistry, Research Center for Analytical Sciences, State Key Laboratory of Medicinal Chemical Biology, and Tianjin Key Laboratory of Molecular Recognition and Biosensing , Nankai University , Tianjin 300071 , China
| |
Collapse
|
32
|
Accumulation of fructose 1,6-bisphosphate protects clear cell renal cell carcinoma from oxidative stress. J Transl Med 2019; 99:898-908. [PMID: 30760861 DOI: 10.1038/s41374-019-0203-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 12/19/2018] [Accepted: 01/04/2019] [Indexed: 01/08/2023] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is characterized by the activation of hypoxia-inducible factors and enhanced aerobic glycolysis. In our previous study, metabolic profiling revealed a threefold increase of fructose 1,6-bisphosphate (FBP) in ccRCC tissue compared with normal kidney tissue. As an important intermediate metabolite, its role in cancer development remains unknown. We found that high levels of FBP were required for cancer growth because of its ability to affect the redox status. Mechanistically, FBP regulated the redox status partially by suppressing NADPH oxidase isoform NOX4 activity in ccRCC cells. ccRCC maintained high levels of FBP through the downregulation of aldolase B (ALDOB). Reduction of FBP levels in cancer cells by the ectopic expression of ALDOB disrupted redox homeostasis, arrested cancer proliferation, and sensitized ccRCC cells to a chemotherapy agent (paclitaxel). Furthermore, low expression of ALDOB portended significantly worse disease-free survival and overall survival in ccRCC patients. In summary, the downregulation of ALDOB and accumulation of FBP promote ccRCC growth by counteracting oxidative stress.
Collapse
|
33
|
Lang L, Chemmalakuzhy R, Shay C, Teng Y. PFKP Signaling at a Glance: An Emerging Mediator of Cancer Cell Metabolism. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1134:243-258. [PMID: 30919341 DOI: 10.1007/978-3-030-12668-1_13] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Phosphofructokinase-1 (PFK-1), a rate-determining enzyme of glycolysis, is an allosteric enzyme that regulates the oxidation of glucose in cellular respiration. Glycolysis phosphofructokinase platelet (PFKP) is the platelet isoform and works as an important mediator of cell metabolism. Considering that PFKP is a crucial player in many steps of cancer initiation and metastasis, we reviewed the specificities and complexities of PFKP and its biological roles in human diseases, especially malignant tumors. The possible use of PFKP as a diagnostic marker or a drug target in the prevention or treatment of cancer is also discussed.
Collapse
Affiliation(s)
- Liwei Lang
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Ron Chemmalakuzhy
- Department of Biology, College of Science and Mathematics, Augusta University, Augusta, GA, USA
| | - Chloe Shay
- The Robinson College of Business, Georgia State University, Atlanta, GA, USA
- Division of Endocrinology and Diabetes, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
| | - Yong Teng
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, USA.
- Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA.
- Department of Medical Laboratory, Imaging and Radiologic Sciences, College of Allied Health, Augusta University, Augusta, GA, USA.
| |
Collapse
|
34
|
Chen G, Liu H, Zhang Y, Liang J, Zhu Y, Zhang M, Yu D, Wang C, Hou J. Silencing PFKP inhibits starvation-induced autophagy, glycolysis, and epithelial mesenchymal transition in oral squamous cell carcinoma. Exp Cell Res 2018; 370:46-57. [DOI: 10.1016/j.yexcr.2018.06.007] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 06/01/2018] [Accepted: 06/09/2018] [Indexed: 01/08/2023]
|
35
|
Xu XR, Yousef GM, Ni H. Cancer and platelet crosstalk: opportunities and challenges for aspirin and other antiplatelet agents. Blood 2018. [PMID: 29519806 DOI: 10.1182/blood-2017-05-743187] [Citation(s) in RCA: 228] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Platelets have long been recognized as key players in hemostasis and thrombosis; however, growing evidence suggests that they are also significantly involved in cancer, the second leading cause of mortality worldwide. Preclinical and clinical studies showed that tumorigenesis and metastasis can be promoted by platelets through a wide variety of crosstalk between platelets and cancer cells. For example, cancer changes platelet behavior by directly inducing tumor-platelet aggregates, triggering platelet granule and extracellular vesicle release, altering platelet phenotype and platelet RNA profiles, and enhancing thrombopoiesis. Reciprocally, platelets reinforce tumor growth with proliferation signals, antiapoptotic effect, and angiogenic factors. Platelets also activate tumor invasion and sustain metastasis via inducing an invasive epithelial-mesenchymal transition phenotype of tumor cells, promoting tumor survival in circulation, tumor arrest at the endothelium, and extravasation. Furthermore, platelets assist tumors in evading immune destruction. Hence, cancer cells and platelets maintain a complex, bidirectional communication. Recently, aspirin (acetylsalicylic acid) has been recognized as a promising cancer-preventive agent. It is recommended at daily low dose by the US Preventive Services Task Force for primary prevention of colorectal cancer. The exact mechanisms of action of aspirin in chemoprevention are not very clear, but evidence has emerged that suggests a platelet-mediated effect. In this article, we will introduce how cancer changes platelets to be more cancer-friendly and highlight advances in the modes of action for aspirin in cancer prevention. We also discuss the opportunities, challenges, and opposing viewpoints on applying aspirin and other antiplatelet agents for cancer prevention and treatment.
Collapse
Affiliation(s)
- Xiaohong Ruby Xu
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, and
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
| | - George M Yousef
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, and
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Heyu Ni
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, and
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Canadian Blood Services Centre for Innovation, Toronto, ON, Canada; and
- Department of Medicine and
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
36
|
Wang J, Tan M, Ge J, Zhang P, Zhong J, Tao L, Wang Q, Tong X, Qiu J. Lysosomal acid lipase promotes cholesterol ester metabolism and drives clear cell renal cell carcinoma progression. Cell Prolif 2018; 51:e12452. [PMID: 29569766 DOI: 10.1111/cpr.12452] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 01/22/2018] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVES Clear cell renal cell carcinoma (ccRCC) is characterized histologically by accumulation of cholesterol esters, cholesterol and other neutral lipids. Lysosomal acid lipase (LAL) is a critical enzyme involved in the cholesterol ester metabolism. Here, we sought to determine whether LAL could orchestrate metabolism of cholesterol esters in order to promote ccRCC progression. MATERIALS AND METHODS Quantitative reverse-transcription PCR and western blots were conducted to assess the expression of LAL in human ccRCC tissues. We analysed the relationship between LAL levels and patient survival using tissue microarrays. We used cell proliferation assays, colony formation assays, cell death assays, metabolic assays and xenograft tumour models to evaluate the biological function and underlying mechanisms. RESULTS LAL was up-regulated in ccRCC tissue. Tissue microarray analysis revealed higher levels of LAL in advanced grades of ccRCC, and high LAL expression indicated lower patient survival. Suppressing LAL expression not only blocked the utilization of cholesterol esters but also impaired proliferation and cellular survival. Furthermore, immunohistochemistry staining showed that LAL expression was correlated with Akt phosphorylation. Suppressing LAL expression decreased the phosphorylation level of Akt and Src and reduced the level of 14,15-epoxyeicosatrienoic acids in ccRCC cells. Supplement of 14,15-epoxyeicosatrienoic acids rescued proliferation in vitro and in vivo. CONCLUSIONS LAL promoted cell proliferation and survival via metabolism of epoxyeicosatrienoic acids and activation of the Src/Akt pathway.
Collapse
Affiliation(s)
- Jun Wang
- Department of Urology, School of Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Mingyue Tan
- Department of Urology, School of Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jifu Ge
- Department of Urology, School of Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ping Zhang
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Zhong
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Le Tao
- Department of Urology, School of Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Qiong Wang
- Department of Clinical Laboratory, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, China
| | - Xuemei Tong
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianxin Qiu
- Department of Urology, School of Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
37
|
Ran H, Zhu Y, Deng R, Zhang Q, Liu X, Feng M, Zhong J, Lin S, Tong X, Su Q. Stearoyl-CoA desaturase-1 promotes colorectal cancer metastasis in response to glucose by suppressing PTEN. J Exp Clin Cancer Res 2018; 37:54. [PMID: 29530061 PMCID: PMC5848567 DOI: 10.1186/s13046-018-0711-9] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 02/14/2018] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Diabetic patients have a higher risk factor for colorectal cancer (CRC) metastasis. Stearoyl-CoA desaturase 1 (SCD1), the main enzyme responsible for producing monounsaturated fatty acids(MUFA) from saturated fatty acids, is frequently deregulated in both diabetes and CRC. The function and mechanism of SCD1 in metastasis of CRC and its relevance to glucose remains largely unknown. METHODS SCD1 expression levels were analyzed in human CRC tissues and the Cancer Browser database ( https://genome-cancer.ucsc.edu/ ). CRC cell lines stably transfected with SCD1 shRNAs or vector were established to investigate the role of SCD1 in modulating migration and invasion of CRC cells. A glucose concentration gradient was set to investigate regulation of SCD1 in CRC relevant to diabetic conditions. RESULTS The clinical data analysis showed high expression of SCD1 in CRC tissues with a negative correlation with the prognosis of CRC. In vitro experiments revealed that SCD1 increased CRC progression through promoting epithelial-mesenchymal transition (EMT). Lipidomic analysis demonstrated that SCD1 increased MUFA levels and MUFA administration could rescue migration and invasion defect of CRC cells induced by SCD1 knockdown. Furthermore, SCD1-mediated progression of CRC was promoted by carbohydrate response-element binding protein (ChREBP) in response to high glucose. Mechanistically, hyperglycemia-SCD1-MUFA induced CRC cell migration and invasion by regulating PTEN. CONCLUSIONS Our findings show that SCD1 promotes metastasis of CRC cells through MUFA production and suppressing PTEN in response to glucose, which may be a novel mechanism for diabetes-induced CRC metastasis.
Collapse
Affiliation(s)
- Hui Ran
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665, Kong Jiang Road, Shanghai, 200092 China
| | - Yemin Zhu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 280 S. Chongqing Road, Shanghai, 200025 China
| | - Ruyuan Deng
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665, Kong Jiang Road, Shanghai, 200092 China
| | - Qi Zhang
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665, Kong Jiang Road, Shanghai, 200092 China
| | - Xisheng Liu
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100, Haining Road, Shanghai, 200080 China
| | - Ming Feng
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 280 S. Chongqing Road, Shanghai, 200025 China
| | - Jie Zhong
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 280 S. Chongqing Road, Shanghai, 200025 China
| | - Shuhai Lin
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 280 S. Chongqing Road, Shanghai, 200025 China
| | - Xuemei Tong
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 280 S. Chongqing Road, Shanghai, 200025 China
| | - Qing Su
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665, Kong Jiang Road, Shanghai, 200092 China
| |
Collapse
|
38
|
Xu J, Shao X, Wei Y, Xu F, Wang H. iTRAQ Proteomic Analysis Reveals That Metabolic Pathways Involving Energy Metabolism Are Affected by Tea Tree Oil in Botrytis cinerea. Front Microbiol 2017; 8:1989. [PMID: 29075250 PMCID: PMC5643485 DOI: 10.3389/fmicb.2017.01989] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 09/27/2017] [Indexed: 11/18/2022] Open
Abstract
Tea tree oil (TTO) is a volatile essential oil obtained from the leaves of the Australian tree Melaleuca alternifolia by vapor distillation. Previously, we demonstrated that TTO has a strong inhibitory effect on Botrytis cinerea. This study investigates the underlying antifungal mechanisms at the molecular level. A proteomics approach using isobaric tags for relative and absolute quantification (iTRAQ) was adopted to investigate the effects of TTO on B. cinerea. A total of 718 differentially expression proteins (DEPs) were identified in TTO-treated samples, 17 were markedly up-regulated and 701 were significantly down-regulated. Among the 718 DEPs, 562 were annotated and classified into 30 functional groups by GO (gene ontology) analysis. KEGG (Kyoto Encyclopedia of Genes and Genomes) enrichment analysis linked 562 DEPs to 133 different biochemical pathways, involving glycolysis, the tricarboxylic acid cycle (TCA cycle), and purine metabolism. Additional experiments indicated that TTO destroys cell membranes and decreases the activities of three enzymes related to the TCA cycle. Our results suggest that TTO treatment inhibits glycolysis, disrupts the TCA cycle, and induces mitochondrial dysfunction, thereby disrupting energy metabolism. This study provides new insights into the mechanisms underlying the antifungal activity of essential oils.
Collapse
Affiliation(s)
- Jiayu Xu
- Department of Food Science and Engineering, Ningbo University, Ningbo, China
| | - Xingfeng Shao
- Department of Food Science and Engineering, Ningbo University, Ningbo, China
| | - Yingying Wei
- Department of Food Science and Engineering, Ningbo University, Ningbo, China
| | - Feng Xu
- Department of Food Science and Engineering, Ningbo University, Ningbo, China
| | - Hongfei Wang
- Department of Food Science and Engineering, Ningbo University, Ningbo, China
| |
Collapse
|
39
|
Cervantes-Anaya N, Ponciano-Gómez A, López-Álvarez GS, Gonzalez-Reyes C, Hernández-Garcia S, Cabañas-Cortes MA, Garrido-Guerrero JE, Villa-Treviño S. Downregulation of sorting nexin 10 is associated with overexpression of miR-30d during liver cancer progression in rats. Tumour Biol 2017; 39:1010428317695932. [PMID: 28381192 DOI: 10.1177/1010428317695932] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
As of 2012, liver cancer was the second leading cause of death worldwide, and hepatocellular carcinoma is the most common primary cancer of the liver. The identification of molecules that might be molecular markers or therapeutic targets is urgently needed to improve clinical management. Based on a microarray analysis performed in our laboratory, we selected six genes-namely, ANXA2, DYNLT1, PFKP, PLA2G7, KRT19, and SNX10-as candidates for validation as tumor markers of liver cancer in a rat model. Their patterns of overexpression in preneoplastic lesions and established tumors at 10 different time points between 24 h and 18 months were analyzed to identify putative tumor markers for further studies. We validated the microarray results by quantitative reverse transcription polymerase chain reaction, which revealed high transcriptional expression for five of the genes, consistent with their high protein expression during cancer progression reported in the literature. However, studies of the association of sorting nexin 10 with different types of cancer are limited, prompting further study. The characterization of sorting nexin 10 in preneoplastic lesions and established tumors revealed messenger RNA overexpression and a simultaneous decrease in sorting nexin 10 protein expression. A group of microRNAs related to sorting nexin 10 messenger RNA were selected based on a data analysis conducted using miRDB and microrna.org . An analysis of the expression of these microRNAs revealed an increase in the transcription of microRNA-30d whenever the sorting nexin 10 protein was downregulated. These results suggest that sorting nexin 10 is a potential liver cancer marker exhibiting characteristics of a putative suppressor protein that is likely regulated by microRNA-30d.
Collapse
Affiliation(s)
- Nancy Cervantes-Anaya
- 1 Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados (CINVESTAV-IPN), Ciudad de México, México
| | - Alberto Ponciano-Gómez
- 2 Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados (CINVESTAV-IPN), Ciudad de México, México
| | - Guadalupe Soledad López-Álvarez
- 1 Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados (CINVESTAV-IPN), Ciudad de México, México
| | - Christian Gonzalez-Reyes
- 1 Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados (CINVESTAV-IPN), Ciudad de México, México.,3 Unidad Académica de Ciencias Químico Biológicas y Farmacéutica, Universidad Autónoma de Nayarit, Tepic, México
| | - Sergio Hernández-Garcia
- 1 Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados (CINVESTAV-IPN), Ciudad de México, México
| | - María Asunción Cabañas-Cortes
- 1 Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados (CINVESTAV-IPN), Ciudad de México, México
| | - José Efraín Garrido-Guerrero
- 2 Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados (CINVESTAV-IPN), Ciudad de México, México
| | - Saúl Villa-Treviño
- 1 Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados (CINVESTAV-IPN), Ciudad de México, México
| |
Collapse
|