151
|
Zhu S, Dong Z, Ke X, Hou J, Zhao E, Zhang K, Wang F, Yang L, Xiang Z, Cui H. The roles of sirtuins family in cell metabolism during tumor development. Semin Cancer Biol 2019; 57:59-71. [DOI: 10.1016/j.semcancer.2018.11.003] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 11/11/2018] [Accepted: 11/14/2018] [Indexed: 12/20/2022]
|
152
|
Li Q, Wang Y, Wu S, Zhou Z, Ding X, Shi R, Thorne RF, Zhang XD, Hu W, Wu M. CircACC1 Regulates Assembly and Activation of AMPK Complex under Metabolic Stress. Cell Metab 2019; 30:157-173.e7. [PMID: 31155494 DOI: 10.1016/j.cmet.2019.05.009] [Citation(s) in RCA: 221] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 03/11/2019] [Accepted: 05/06/2019] [Indexed: 12/15/2022]
Abstract
We report that circACC1, a circular RNA derived from human ACC1, plays a critical role in cellular responses to metabolic stress. CircACC1 is preferentially produced over ACC1 in response to serum deprivation by the transcription factor c-Jun. It functions to stabilize and promote the enzymatic activity of the AMPK holoenzyme by forming a ternary complex with the regulatory β and γ subunits. The cellular levels of circACC1 modulate both fatty acid β-oxidation and glycolysis, resulting in profound changes in cellular lipid storage. In a tumor xenograft model, silencing or enforced expression of circACC1 resulted in growth inhibition and enhancement, respectively. Moreover, increased AMPK activation in colorectal cancer tissues was frequently associated with elevated circACC1 expression. We conclude that circACC1 serves as an economic means to elicit AMPK activation and moreover propose that cancer cells exploit circACC1 during metabolic reprogramming.
Collapse
Affiliation(s)
- Qidong Li
- The Chinese Academy of Sciences (CAS), Key Laboratory of Innate Immunity & Chronic Disease, CAS Center for Excellence in Cell & Molecular Biology, School of Life Sciences, University of Science & Technology of China, Hefei 230026, China
| | - Yichun Wang
- The Chinese Academy of Sciences (CAS), Key Laboratory of Innate Immunity & Chronic Disease, CAS Center for Excellence in Cell & Molecular Biology, School of Life Sciences, University of Science & Technology of China, Hefei 230026, China
| | - Shuang Wu
- Department of Immunology, Anhui Medical University, Hefei 230027, China
| | - Zhong Zhou
- The Chinese Academy of Sciences (CAS), Key Laboratory of Innate Immunity & Chronic Disease, CAS Center for Excellence in Cell & Molecular Biology, School of Life Sciences, University of Science & Technology of China, Hefei 230026, China
| | - Xiaojuan Ding
- Department of Immunology, Anhui Medical University, Hefei 230027, China
| | - Ronghua Shi
- The Chinese Academy of Sciences (CAS), Key Laboratory of Innate Immunity & Chronic Disease, CAS Center for Excellence in Cell & Molecular Biology, School of Life Sciences, University of Science & Technology of China, Hefei 230026, China
| | - Rick F Thorne
- Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical Science, Zhengzhou University, Zhengzhou 450003, China; Key Laboratory of Stem Cell Differentiation & Modification, School of Clinical Medicine, Henan University, Zhengzhou 450003, China; School of Environmental & Life Sciences, University of Newcastle, Newcastle, NSW 2258, Australia
| | - Xu Dong Zhang
- Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical Science, Zhengzhou University, Zhengzhou 450003, China; Key Laboratory of Stem Cell Differentiation & Modification, School of Clinical Medicine, Henan University, Zhengzhou 450003, China; School of Biomedical Sciences & Pharmacy, University of Newcastle, Newcastle, NSW 2308, Australia
| | - Wanglai Hu
- Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical Science, Zhengzhou University, Zhengzhou 450003, China; Key Laboratory of Stem Cell Differentiation & Modification, School of Clinical Medicine, Henan University, Zhengzhou 450003, China; Department of Immunology, Anhui Medical University, Hefei 230027, China.
| | - Mian Wu
- The Chinese Academy of Sciences (CAS), Key Laboratory of Innate Immunity & Chronic Disease, CAS Center for Excellence in Cell & Molecular Biology, School of Life Sciences, University of Science & Technology of China, Hefei 230026, China; Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical Science, Zhengzhou University, Zhengzhou 450003, China; Key Laboratory of Stem Cell Differentiation & Modification, School of Clinical Medicine, Henan University, Zhengzhou 450003, China.
| |
Collapse
|
153
|
Wnt Signaling in Cancer Metabolism and Immunity. Cancers (Basel) 2019; 11:cancers11070904. [PMID: 31261718 PMCID: PMC6678221 DOI: 10.3390/cancers11070904] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 06/22/2019] [Accepted: 06/26/2019] [Indexed: 12/14/2022] Open
Abstract
The Wingless (Wnt)/β-catenin pathway has long been associated with tumorigenesis, tumor plasticity, and tumor-initiating cells called cancer stem cells (CSCs). Wnt signaling has recently been implicated in the metabolic reprogramming of cancer cells. Aberrant Wnt signaling is considered to be a driver of metabolic alterations of glycolysis, glutaminolysis, and lipogenesis, processes essential to the survival of bulk and CSC populations. Over the past decade, the Wnt pathway has also been shown to regulate the tumor microenvironment (TME) and anti-cancer immunity. Wnt ligands released by tumor cells in the TME facilitate the immune evasion of cancer cells and hamper immunotherapy. In this review, we illustrate the role of the canonical Wnt/β-catenin pathway in cancer metabolism and immunity to explore the potential therapeutic approach of targeting Wnt signaling from a metabolic and immunological perspective.
Collapse
|
154
|
Do SK, Choi SH, Lee SY, Choi JE, Hong MJ, Kang HG, Lee WK, Lee EB, Shin KM, Jeong JY, Lee YH, Seo H, Yoo SS, Lee J, Cha SI, Kim CH, Seok Y, Cho S, Jheon S, Park JY. Glucose transporter 3 gene variant is associated with survival outcome of patients with non-small cell lung cancer after surgical resection. Gene 2019; 703:58-64. [DOI: 10.1016/j.gene.2019.04.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 03/20/2019] [Accepted: 04/03/2019] [Indexed: 02/04/2023]
|
155
|
Arif T, Amsalem Z, Shoshan-Barmatz V. Metabolic Reprograming Via Silencing of Mitochondrial VDAC1 Expression Encourages Differentiation of Cancer Cells. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 17:24-37. [PMID: 31195298 PMCID: PMC6562189 DOI: 10.1016/j.omtn.2019.05.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 05/09/2019] [Accepted: 05/09/2019] [Indexed: 12/29/2022]
Abstract
The mitochondrial gatekeeper voltage-dependent anion channel 1 (VDAC1) controls metabolic and energy cross-talk between mitochondria and the rest of the cell and is involved in mitochondria-mediated apoptosis. Here, we compared the effects of downregulated VDAC1 expression in the U-87MG glioblastoma, MDA-MB-231 triple-negative breast cancer (TNBC), and A549 lung cancer cell lines, using small interfering RNA (siRNA) specific to human VDAC1 (si-hVDAC1). The cells were subjected to si-hVDAC1 (50 nM) treatment for 5–20 days. Although VDAC1 silencing occurred within a day, the cells underwent reprograming with respect to rewiring metabolism, elimination of cancer stem cells (CSCs), and alteration of transcription factor (TF) expression and proteins associated with differentiation, with maximal changes being observed after 3 weeks of silencing VDAC1 expression. The differentiation into fewer tumorigenic cells may be associated with the elimination of CSCs. These alterations are interconnected, as protein up- or downregulation occurred simultaneously, starting 15–20 days after VDAC1 levels were first decreased. Moreover, the VDAC1 depletion-mediated effects on a network of key regulators of cell metabolism, CSCs, TFs, and other factors leading to differentiation are coordinated and are common to the glioblastoma multiforme (GBM) and lung and breast cancer cell lines, despite differing in origin and carried mutations. Thus, our study showed that VDAC1 depletion triggers reprograming of malignant cancer cells into terminally differentiated cells and that this may be a promising therapeutic approach for various cancers.
Collapse
Affiliation(s)
- Tasleem Arif
- Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Zohar Amsalem
- Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Varda Shoshan-Barmatz
- Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel.
| |
Collapse
|
156
|
Orang AV, Petersen J, McKinnon RA, Michael MZ. Micromanaging aerobic respiration and glycolysis in cancer cells. Mol Metab 2019; 23:98-126. [PMID: 30837197 PMCID: PMC6479761 DOI: 10.1016/j.molmet.2019.01.014] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 01/22/2019] [Accepted: 01/30/2019] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Cancer cells possess a common metabolic phenotype, rewiring their metabolic pathways from mitochondrial oxidative phosphorylation to aerobic glycolysis and anabolic circuits, to support the energetic and biosynthetic requirements of continuous proliferation and migration. While, over the past decade, molecular and cellular studies have clearly highlighted the association of oncogenes and tumor suppressors with cancer-associated glycolysis, more recent attention has focused on the role of microRNAs (miRNAs) in mediating this metabolic shift. Accumulating studies have connected aberrant expression of miRNAs with direct and indirect regulation of aerobic glycolysis and associated pathways. SCOPE OF REVIEW This review discusses the underlying mechanisms of metabolic reprogramming in cancer cells and provides arguments that the earlier paradigm of cancer glycolysis needs to be updated to a broader concept, which involves interconnecting biological pathways that include miRNA-mediated regulation of metabolism. For these reasons and in light of recent knowledge, we illustrate the relationships between metabolic pathways in cancer cells. We further summarize our current understanding of the interplay between miRNAs and these metabolic pathways. This review aims to highlight important metabolism-associated molecular components in the hunt for selective preventive and therapeutic treatments. MAJOR CONCLUSIONS Metabolism in cancer cells is influenced by driver mutations but is also regulated by posttranscriptional gene silencing. Understanding the nuanced regulation of gene expression in these cells and distinguishing rapid cellular responses from chronic adaptive mechanisms provides a basis for rational drug design and novel therapeutic strategies.
Collapse
Affiliation(s)
- Ayla V Orang
- Flinders Centre for Innovation in Cancer, Flinders University, Flinders Medical Centre, Adelaide, South Australia 5042, Australia.
| | - Janni Petersen
- Flinders Centre for Innovation in Cancer, Flinders University, Flinders Medical Centre, Adelaide, South Australia 5042, Australia.
| | - Ross A McKinnon
- Flinders Centre for Innovation in Cancer, Flinders University, Flinders Medical Centre, Adelaide, South Australia 5042, Australia.
| | - Michael Z Michael
- Flinders Centre for Innovation in Cancer, Flinders University, Flinders Medical Centre, Adelaide, South Australia 5042, Australia.
| |
Collapse
|
157
|
Marsolier J, Perichon M, Weitzman JB, Medjkane S. Secreted parasite Pin1 isomerase stabilizes host PKM2 to reprogram host cell metabolism. Commun Biol 2019; 2:152. [PMID: 31044177 PMCID: PMC6491484 DOI: 10.1038/s42003-019-0386-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 03/05/2019] [Indexed: 01/04/2023] Open
Abstract
Metabolic reprogramming is an important feature of host-pathogen interactions and a hallmark of tumorigenesis. The intracellular apicomplexa parasite Theileria induces a Warburg-like effect in host leukocytes by hijacking signaling machineries, epigenetic regulators and transcriptional programs to create a transformed cell state. The molecular mechanisms underlying host cell transformation are unclear. Here we show that a parasite-encoded prolyl-isomerase, TaPin1, stabilizes host pyruvate kinase isoform M2 (PKM2) leading to HIF-1α-dependent regulation of metabolic enzymes, glucose uptake and transformed phenotypes in parasite-infected cells. Our results provide a direct molecular link between the secreted parasite TaPin1 protein and host gene expression programs. This study demonstrates the importance of prolyl isomerization in the parasite manipulation of host metabolism.
Collapse
Affiliation(s)
- Justine Marsolier
- Sorbonne Paris Cité, Epigenetics and Cell Fate, Université Paris Diderot, CNRS, UMR 7216 Paris, France
- Present Address: Institut Curie, 26 rue d′Ulm, 75005 Paris, France
| | - Martine Perichon
- Sorbonne Paris Cité, Epigenetics and Cell Fate, Université Paris Diderot, CNRS, UMR 7216 Paris, France
| | - Jonathan B. Weitzman
- Sorbonne Paris Cité, Epigenetics and Cell Fate, Université Paris Diderot, CNRS, UMR 7216 Paris, France
| | - Souhila Medjkane
- Sorbonne Paris Cité, Epigenetics and Cell Fate, Université Paris Diderot, CNRS, UMR 7216 Paris, France
| |
Collapse
|
158
|
B7-H3 promotes aerobic glycolysis and chemoresistance in colorectal cancer cells by regulating HK2. Cell Death Dis 2019; 10:308. [PMID: 30952834 PMCID: PMC6450969 DOI: 10.1038/s41419-019-1549-6] [Citation(s) in RCA: 170] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/22/2019] [Accepted: 03/25/2019] [Indexed: 02/06/2023]
Abstract
Accumulating evidence suggests that aerobic glycolysis is important for colorectal cancer (CRC) development. However, the underlying mechanisms have yet to be elucidated. B7-H3, an immunoregulatory protein, is broadly overexpressed by multiple tumor types and plays a vital role in tumor progression. In this study, we found that overexpression of B7-H3 effectively increased the rate of glucose consumption and lactate production, whereas knockdown of B7-H3 had the opposite effect. Furthermore, we showed that B7-H3 increased glucose consumption and lactate production by promoting hexokinase 2 (HK2) expression in CRC cells, and we also found that HK2 was a key mediator of B7-H3-induced CRC chemoresistance. Depletion of HK2 expression or treating cells with HK2 inhibitors could reverse the B7-H3-induced increase in aerobic glycolysis and B7-H3-endowed chemoresistance of cancer cells. Moreover, we verified a positive correlation between the expression of B7-H3 and HK2 in tumor tissues of CRC patients. Collectively, our findings suggest that B7-H3 may be a novel regulator of glucose metabolism and chemoresistance via controlling HK2 expression in CRC cells, a result that could help develop B7-H3 as a promising therapeutic target for CRC treatment.
Collapse
|
159
|
Liu P, Zhu L, Zhang F, Lin J, Du M, Cao Z, Ma L, Hu Z. LncRNA UCA1/miR-143 miR-216b/HK2/MAPK signaling pathway is involved in the regulation of endothelial cell proliferation via the modulation of glycolysis in melanoma. EUR J INFLAMM 2019. [DOI: 10.1177/2058739219837050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) and microRNAs (miRNAs/miRs) are noncoding RNAs that function as regulators of tumor suppressors and oncogenes. The aim of the present study was to investigate the potential mechanism associated with the involvement of urothelial cancer associated 1 (UCA1) in melanoma. Reverse transcription-quantitative polymerase chain reaction and western blot analysis were performed in order to determine the expression levels of UCA1, miR-143, miR-216b, and hexokinase 2 (HK2) in the melanoma and control groups, as well as the influence of UCA1, miR-143, and miR-216b on the expression of HK2, and the effect of lactate and UCA1 on the phosphorylation of p38 mitogen-activated protein kinase (p38 MAPK). Bioinformatics algorithm analysis and a luciferase assay were performed in order to predict miRNA targets. In addition, an MTT assay was performed in order to determine the effect of lactate and UCA1 expression on cell proliferation. A total of 39 participants, consisting of 18 patients with melanoma and 21 healthy control subjects, were included in the present study. The present study demonstrated that the expression levels of UCA1 mRNA, and HK2 mRNA and protein were enhanced in patients with melanoma compared with healthy controls; whereas the expression levels of miR-143 and miR-216b mRNA were suppressed in patients with melanoma compared with healthy controls. Furthermore, it was revealed that UCA1 negatively modulated the expression of miR-143 and miR-216b, and that miR-143 and miR-216b directly targeted the HK2 protein by binding to the HK2 3′ untranslated region (UTR). In addition, it was demonstrated that miR-143 and miR-216 suppressed the luciferase activity exhibited by wild-type HK2 3′-UTR. Furthermore, it was revealed that transfection with UCA1 small interfering RNA, and miR-143 and miR-216b mimics markedly suppressed HK2 mRNA and protein expression levels as well as lactate levels in human umbilical vein endothelial cells; however, O2 consumption was revealed to be enhanced post transfection. By contrast, transfection with UCA1 enhanced HK2 mRNA and protein expression levels as well as lactate production; however, O2 consumption was revealed to be suppressed post transfection. Lactate-induced phosphorylation of p38 MAPK was revealed to occur in a concentration-dependent manner, and UCA1 enhanced the phosphorylation level of p38 MAPK via the inhibition of miR-143 and miR-216b expression. Lactate and UCA1 were demonstrated to enhance cell proliferation. In conclusion, the present study demonstrated that the lncRNA UCA1/miR-143 miR-216b/HK2/lactic acid/MAPK axis may be involved in the pathogenesis of melanoma via the modulation of endothelial cells, and thus, lncRNA UCA1 may serve as a potential therapeutic target for melanoma treatment.
Collapse
Affiliation(s)
- Pei Liu
- Department of Plastic Surgery, Qilu Hospital of Shandong University, Jinan, P.R. China
| | - Lei Zhu
- Department of Hand and Foot Surgery, Qilu Hospital of Shandong University, Jinan, P.R. China
| | - Fan Zhang
- Department of Plastic Surgery, Qilu Hospital of Shandong University, Jinan, P.R. China
| | - Junhao Lin
- Department of Plastic Surgery, Qilu Hospital of Shandong University, Jinan, P.R. China
- Department of Hand and Foot Surgery, Qilu Hospital of Shandong University, Jinan, P.R. China
| | - Min Du
- Department of Plastic Surgery, Qilu Hospital of Shandong University, Jinan, P.R. China
| | - Zilong Cao
- Department of Plastic Surgery, Qilu Hospital of Shandong University, Jinan, P.R. China
| | - Ling Ma
- Department of Plastic Surgery, Qilu Hospital of Shandong University, Jinan, P.R. China
| | - Zhensheng Hu
- Department of Plastic Surgery, Qilu Hospital of Shandong University, Jinan, P.R. China
| |
Collapse
|
160
|
Yu W, Liu T, Zhang M, Wang Z, Ye J, Li CX, Liu W, Li R, Feng J, Zhang XZ. O 2 Economizer for Inhibiting Cell Respiration To Combat the Hypoxia Obstacle in Tumor Treatments. ACS NANO 2019; 13:1784-1794. [PMID: 30698953 DOI: 10.1021/acsnano.8b07852] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Hypoxia, a ubiquitously aberrant phenomenon implicated in tumor growth, causes severe tumor resistance to therapeutic interventions. Instead of the currently prevalent solution through intratumoral oxygen supply, we put forward an "O2-economizer" concept by inhibiting the O2 consumption of cell respiration to spare endogenous O2 and overcome the hypoxia barrier. A nitric oxide (NO) donor responsible for respiration inhibition and a photosensitizer for photodynamic therapy (PDT) are co-loaded into poly(d,l-lactide- co-glycolide) nanovesicles to provide a PDT-specific O2 economizer. Once accumulating in tumors and subsequently responding to the locally reductive environment, the carried NO donor undergoes breakdown to produce NO for inhibiting cellular respiration, allowing more O2 in tumor cells to support the profound enhancement of PDT. Depending on the biochemical reallocation of cellular oxygen resource, this O2-economizer concept offers a way to address the important issue of hypoxia-induced tumor resistance to therapeutic interventions, including but not limited to PDT.
Collapse
Affiliation(s)
- Wuyang Yu
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry , Wuhan University , Wuhan 430072 , PR China
| | - Tao Liu
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry , Wuhan University , Wuhan 430072 , PR China
| | - Mingkang Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry , Wuhan University , Wuhan 430072 , PR China
| | - Zixu Wang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry , Wuhan University , Wuhan 430072 , PR China
| | - Jingjie Ye
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry , Wuhan University , Wuhan 430072 , PR China
| | - Chu-Xin Li
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry , Wuhan University , Wuhan 430072 , PR China
| | - Wenlong Liu
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry , Wuhan University , Wuhan 430072 , PR China
| | - Runqing Li
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry , Wuhan University , Wuhan 430072 , PR China
| | - Jun Feng
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry , Wuhan University , Wuhan 430072 , PR China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry , Wuhan University , Wuhan 430072 , PR China
| |
Collapse
|
161
|
Zhao S, Feng J, Li C, Gao H, Lv P, Li J, Liu Q, He Y, Wang H, Gong L, Li D, Zhang Y. Phosphoproteome profiling revealed abnormally phosphorylated AMPK and ATF2 involved in glucose metabolism and tumorigenesis of GH-PAs. J Endocrinol Invest 2019; 42:137-148. [PMID: 29691806 DOI: 10.1007/s40618-018-0890-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 04/11/2018] [Indexed: 01/04/2023]
Abstract
PURPOSE Protein phosphorylation plays a key role in tumorigenesis and progression. However, little is known about the phosphoproteome profiles of growth hormone-secreting pituitary adenomas (GH-PAs). The aim of this study was to identify critical biomarkers and signaling pathways that might play important roles in GH-PAs and may, therefore, represent potential therapeutic targets. METHODS The differential phosphoprotein expression patterns involved in GH-PAs were investigated by nano-LC-MS/MS in a group of samples. The phosphoprotein expression data were analyzed by bioinformatics. The expression levels of the candidate phosphorylated AMPK (ser496) and ATF2 (ser112) were validated by Western blot analysis in another group of samples. RESULTS A total of 1213 phosphorylated protein sites corresponding to 667 proteins were significantly different between GH-PAs and healthy pituitary glands. Among these phosphorylated sites, 871 exhibited lower levels of phosphorylation in GH-PAs. Moreover, 140 novel phosphosites corresponding to 93 proteins were differentially phosphorylated between GH-PAs and healthy pituitary glands, 101 of which showed decreased phosphorylation in GH-PAs. The majority of differentially expressed phosphorylated proteins were significantly enriched in glycolysis and the AMPK signaling pathway in GH-PAs. The AMPK signaling pathway was demonstrated to be inhibited in GH-PAs by pathway activity analysis (z score = - 2.324). Notably, the phosphorylated levels of AMPK (ser496) and ATF2 (ser112) were significantly lower in GH-PAs than in healthy pituitary glands. CONCLUSION These findings suggest that decreased phosphorylation of the AMPK/ATF2 pathway may be critical for glucose metabolism and tumorigenesis in GH-PAs.
Collapse
Affiliation(s)
- S Zhao
- Beijing Neurosurgical Institute, Capital Medical University, TianTanXiLi6, Beijing, 100050, China.
| | - J Feng
- Beijing Neurosurgical Institute, Capital Medical University, TianTanXiLi6, Beijing, 100050, China
| | - C Li
- Beijing Neurosurgical Institute, Capital Medical University, TianTanXiLi6, Beijing, 100050, China
| | - H Gao
- Beijing Neurosurgical Institute, Capital Medical University, TianTanXiLi6, Beijing, 100050, China
| | - P Lv
- Beijing Neurosurgical Institute, Capital Medical University, TianTanXiLi6, Beijing, 100050, China
- Chinese Medical Association, Beijing, 100710, China
| | - J Li
- Beijing Neurosurgical Institute, Capital Medical University, TianTanXiLi6, Beijing, 100050, China
| | - Q Liu
- Beijing Neurosurgical Institute, Capital Medical University, TianTanXiLi6, Beijing, 100050, China
| | - Y He
- Beijing Neurosurgical Institute, Capital Medical University, TianTanXiLi6, Beijing, 100050, China
| | - H Wang
- Beijing Neurosurgical Institute, Capital Medical University, TianTanXiLi6, Beijing, 100050, China
| | - L Gong
- Beijing Neurosurgical Institute, Capital Medical University, TianTanXiLi6, Beijing, 100050, China
| | - D Li
- Beijing Neurosurgical Institute, Capital Medical University, TianTanXiLi6, Beijing, 100050, China
| | - Y Zhang
- Beijing Neurosurgical Institute, Capital Medical University, TianTanXiLi6, Beijing, 100050, China.
- Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050, China.
- Beijing Institute for Brain Disorders Brain Tumor Center, Capital Medical University, Beijing, 100050, China.
- China National Clinical Research Center for Neurological Diseases, Beijing, 100050, China.
| |
Collapse
|
162
|
Kouvaras E, Christoni Z, Siasios I, Malizos K, Koukoulis GK, Ioannou M. Hypoxia-inducible factor 1-alpha and vascular endothelial growth factor in cartilage tumors. Biotech Histochem 2019; 94:283-289. [PMID: 30605014 DOI: 10.1080/10520295.2018.1556806] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Neoangiogenesis has been demonstrated in chondrosarcoma. Anti-angiogenic therapies are being tested in clinical trials for chondrosarcomas. Studies of the underlying mechanisms have been performed almost exclusively in cell lines. We immunostained 20 samples of chondrosarcoma and 20 samples of enchondromas with antibodies against hypoxia-inducible factor 1-alpha (HIF-1-alpha) and vascular endothelial growth factor (VEGF). The immunohistochemical staining of HIF-1-alpha and VEGF were highly correlated. Enchondromas were HIF-1-alpha and VEGF negative, whereas all chondrosarcoma exhibited HIF-1-alpha and VEGF immunostaining. HIF-1-alpha/VEGF double positive cases were almost exclusively chondrosarcomas with a high tumor grade. We suggest that HIF-1-alpha is a marker of malignancy in chondrosarcomas that correlates with tumor neo-angiogenesis. Our findings also suggest that a HIF-1-alpha/VEGF angiogenic pathway may exist in chondrosarcoma in vivo as in other malignant tumors. The inclusion of novel inhibitors to HIF-1-alpha and other factors may optimize anti-angiogenic interventions in chondrosarcoma.
Collapse
Affiliation(s)
- E Kouvaras
- a Department of Pathology , Medical School, University of Thessaly , Larisa , Greece
| | - Z Christoni
- a Department of Pathology , Medical School, University of Thessaly , Larisa , Greece
| | - I Siasios
- a Department of Pathology , Medical School, University of Thessaly , Larisa , Greece
| | - K Malizos
- b Orthopaedic Department , Medical School, University of Thessaly , Larisa , Greece
| | - G K Koukoulis
- a Department of Pathology , Medical School, University of Thessaly , Larisa , Greece
| | - M Ioannou
- a Department of Pathology , Medical School, University of Thessaly , Larisa , Greece
| |
Collapse
|
163
|
Mitochondrial VDAC1 Silencing Leads to Metabolic Rewiring and the Reprogramming of Tumour Cells into Advanced Differentiated States. Cancers (Basel) 2018; 10:cancers10120499. [PMID: 30544833 PMCID: PMC6316808 DOI: 10.3390/cancers10120499] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 11/27/2018] [Accepted: 12/04/2018] [Indexed: 01/10/2023] Open
Abstract
Oncogenic properties, along with the metabolic reprogramming necessary for tumour growth and motility, are acquired by cancer cells. Thus, tumour metabolism is becoming a target for cancer therapy. Here, cancer cell metabolism was tackled by silencing the expression of voltage-dependent anion channel 1 (VDAC1), a mitochondrial protein that controls cell energy, as well as metabolic and survival pathways and that is often over-expressed in many cancers. We demonstrated that silencing VDAC1 expression using human-specific siRNA (si-hVDAC1) inhibited cancer cell growth, both in vitro and in mouse xenograft models of human glioblastoma (U-87MG), lung cancer (A549), and triple negative breast cancer (MDA-MB-231). Importantly, treatment with si-hVDAC1 induced metabolic rewiring of the cancer cells, reversing their oncogenic properties and diverting them towards differentiated-like cells. The si-hVDAC1-treated residual “tumour” showed reprogrammed metabolism, decreased proliferation, inhibited stemness and altered expression of genes and proteins, leading to cell differentiation toward less malignant lineages. These VDAC1 depletion-mediated effects involved alterations in master transcription factors associated with cancer hallmarks, such as highly increased expression of p53 and decreased expression of HIF-1a and c-Myc that regulate signalling pathways (e.g., AMPK, mTOR). High expression of p53 and the pro-apoptotic proteins cytochrome c and caspases without induction of apoptosis points to functions for these proteins in promoting cell differentiation. These results clearly show that VDAC1 depletion similarly leads to a rewiring of cancer cell metabolism in breast and lung cancer and glioblastoma, regardless of origin or mutational status. This metabolic reprogramming results in cell growth arrest and inhibited tumour growth while encouraging cell differentiation, thus generating cells with decreased proliferation capacity. These results further suggest VDAC1 to be an innovative and markedly potent therapeutic target.
Collapse
|
164
|
Srivastava A, Creek DJ. Discovery and Validation of Clinical Biomarkers of Cancer: A Review Combining Metabolomics and Proteomics. Proteomics 2018; 19:e1700448. [PMID: 30353665 DOI: 10.1002/pmic.201700448] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 10/11/2018] [Indexed: 12/19/2022]
Abstract
Early detection and diagnosis of cancer can allow timely medical intervention, which greatly improves chances of survival and enhances quality of life. Biomarkers play an important role in assisting clinicians and health care providers in cancer diagnosis and treatment follow-up. In spite of years of research and the discovery of thousands of candidate cancer biomarkers, only a few have transitioned to routine usage in the clinic. This review highlights advances in proteomics technologies that have enabled high rates of discovery of candidate cancer biomarkers and evaluates integration with other omics technologies to improve their progress through to validation and clinical translation. Furthermore, it gauges the role of metabolomics technology in cancer biomarker research and assesses it as a complementary tool in aiding cancer biomarker discovery and validation.
Collapse
Affiliation(s)
- Anubhav Srivastava
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria, 3052, Australia
| | - Darren John Creek
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria, 3052, Australia
| |
Collapse
|
165
|
Tamori S, Nozaki Y, Motomura H, Nakane H, Katayama R, Onaga C, Kikuchi E, Shimada N, Suzuki Y, Noike M, Hara Y, Sato K, Sato T, Yamamoto K, Hanawa T, Imai M, Abe R, Yoshimori A, Takasawa R, Tanuma SI, Akimoto K. Glyoxalase 1 gene is highly expressed in basal-like human breast cancers and contributes to survival of ALDH1-positive breast cancer stem cells. Oncotarget 2018; 9:36515-36529. [PMID: 30559934 PMCID: PMC6284866 DOI: 10.18632/oncotarget.26369] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 11/01/2018] [Indexed: 12/13/2022] Open
Abstract
Glyoxalase 1 (GLO1) is a ubiquitous enzyme involved in the detoxification of methylglyoxal, a cytotoxic byproduct of glycolysis that induces apoptosis. In this study, we found that GLO1 gene expression correlates with neoplasm histologic grade (χ 2 test, p = 0.002) and is elevated in human basal-like breast cancer tissues. Approximately 90% of basal-like cancers were grade 3 tumors highly expressing both GLO1 and the cancer stem cell marker ALDH1A3. ALDH1high cells derived from the MDA-MB 157 and MDA-MB 468 human basal-like breast cancer cell lines showed elevated GLO1 activity. GLO1 inhibition using TLSC702 suppressed ALDH1high cell viability as well as the formation of tumor-spheres by ALDH1high cells. GLO1 knockdown using specific siRNAs also suppressed ALDH1high cell viability, and both TLSC702 and GLO1 siRNA induced apoptosis in ALDH1high cells. These results suggest GLO1 is essential for the survival of ALDH1-positive breast cancer stem cells. We therefore conclude that GLO1 is a potential therapeutic target for treatment of basal-like breast cancers.
Collapse
Affiliation(s)
- Shoma Tamori
- Department of Medicinal and Life Science, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
- Translational Research Center, Research Institute for Science and Technology, Tokyo University of Science, Chiba, Japan
| | - Yuka Nozaki
- Department of Medicinal and Life Science, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
- Translational Research Center, Research Institute for Science and Technology, Tokyo University of Science, Chiba, Japan
| | - Hitomi Motomura
- Department of Medicinal and Life Science, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
- Translational Research Center, Research Institute for Science and Technology, Tokyo University of Science, Chiba, Japan
| | - Hiromi Nakane
- Department of Medicinal and Life Science, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Reika Katayama
- Department of Medicinal and Life Science, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Chotaro Onaga
- Department of Medicinal and Life Science, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Eriko Kikuchi
- Department of Medicinal and Life Science, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Nami Shimada
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Yuhei Suzuki
- Department of Medicinal and Life Science, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Mei Noike
- Department of Medicinal and Life Science, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Yasushi Hara
- Research Institute for Biochemical Sciences, Tokyo University of Science, Chiba, Japan
| | - Keiko Sato
- Department of Information Sciences, Faculty of Science and Technology, Tokyo University of Science, Chiba, Japan
- Translational Research Center, Research Institute for Science and Technology, Tokyo University of Science, Chiba, Japan
| | - Tsugumichi Sato
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
- Translational Research Center, Research Institute for Science and Technology, Tokyo University of Science, Chiba, Japan
| | - Kouji Yamamoto
- Department of Biostatistics, Yokohama City University, School of Medicine, Yokohama, Japan
- Translational Research Center, Research Institute for Science and Technology, Tokyo University of Science, Chiba, Japan
| | - Takehisa Hanawa
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
- Translational Research Center, Research Institute for Science and Technology, Tokyo University of Science, Chiba, Japan
| | - Misa Imai
- Department of Hematology, Juntendo University School of Medicine, Tokyo, Japan
- Leading Center for the Development and Research of Cancer Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | - Ryo Abe
- Research Institute for Biochemical Sciences, Tokyo University of Science, Chiba, Japan
- Strategic Innovation and Research Center, Teikyo University, Tokyo, Japan
| | | | - Ryoko Takasawa
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Sei-Ichi Tanuma
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
- Laboratory of Genomic Medicinal Science, Research Institute for Science and Technology, Tokyo University of Science, Chiba, Japan
| | - Kazunori Akimoto
- Department of Medicinal and Life Science, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
- Translational Research Center, Research Institute for Science and Technology, Tokyo University of Science, Chiba, Japan
| |
Collapse
|
166
|
Ippolito L, Morandi A, Giannoni E, Chiarugi P. Lactate: A Metabolic Driver in the Tumour Landscape. Trends Biochem Sci 2018; 44:153-166. [PMID: 30473428 DOI: 10.1016/j.tibs.2018.10.011] [Citation(s) in RCA: 332] [Impact Index Per Article: 47.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 09/14/2018] [Accepted: 10/25/2018] [Indexed: 01/07/2023]
Abstract
The presence of lactate in human tumours has been long neglected, confined to the role of a waste product derived from glycolysis and as a biomarker of malignancy. More recently, lactate has been rediscovered as signalling molecule that plays important roles in the regulation of the metabolic pathways, the immune response, and cell-to-cell communication within the tumour microenvironment. This review examines recent discoveries about the functional role of lactate in shaping the behaviour and the phenotype of tumour and tumour-associated cells, and describes potential clinical approaches to target lactate transport and metabolism in tumours.
Collapse
Affiliation(s)
- Luigi Ippolito
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy
| | - Andrea Morandi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy
| | - Elisa Giannoni
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy
| | - Paola Chiarugi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy.
| |
Collapse
|
167
|
Liao R, Ren G, Liu H, Chen X, Cao Q, Wu X, Li J, Dong C. ME1 promotes basal-like breast cancer progression and associates with poor prognosis. Sci Rep 2018; 8:16743. [PMID: 30425310 PMCID: PMC6233160 DOI: 10.1038/s41598-018-35106-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 06/27/2018] [Indexed: 12/14/2022] Open
Abstract
Basal-like breast cancer (BLBC) is associated with a poor clinical outcome due to the few treatment options and absence of effective targeted agents. Here, we show that malic enzyme 1 (ME1) is dramatically upregulated in BLBC due to ME1 copy number amplification. ME1 expression increases glucose uptake and lactate production, and reduces oxygen consumption, leading to aerobic glycolysis. ME1 expression promotes, whereas knockdown of ME1 expression suppresses tumorigenicity. In breast cancer patients, ME1 expression is positively correlated with large tumor size, high grade, poor survival, and chemotherapy resistance. Our study not only contributes to a new understanding of how metabolic reprogramming contributes to BLBC progression, but also provides a potential prognostic marker and therapeutic target for this challenging disease.
Collapse
Affiliation(s)
- Ruocen Liao
- Department of Pathology and Pathophysiology, and Department of Surgical Oncology (breast center) of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Guoping Ren
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Huixin Liu
- Department of Pathology and Pathophysiology, and Department of Surgical Oncology (breast center) of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Xingyu Chen
- Department of Pathology and Pathophysiology, and Department of Surgical Oncology (breast center) of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Qianhua Cao
- Department of Pathology and Pathophysiology, and Department of Surgical Oncology (breast center) of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Xuebiao Wu
- Department of Pathology and Pathophysiology, and Department of Surgical Oncology (breast center) of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Jun Li
- Department of Pathology and Pathophysiology, and Department of Surgical Oncology (breast center) of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Chenfang Dong
- Department of Pathology and Pathophysiology, and Department of Surgical Oncology (breast center) of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China. .,Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| |
Collapse
|
168
|
Vallée A, Guillevin R, Vallée JN. Vasculogenesis and angiogenesis initiation under normoxic conditions through Wnt/β-catenin pathway in gliomas. Rev Neurosci 2018; 29:71-91. [PMID: 28822229 DOI: 10.1515/revneuro-2017-0032] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 06/25/2017] [Indexed: 12/11/2022]
Abstract
The canonical Wnt/β-catenin pathway is up-regulated in gliomas and involved in proliferation, invasion, apoptosis, vasculogenesis and angiogenesis. Nuclear β-catenin accumulation correlates with malignancy. Hypoxia activates hypoxia-inducible factor (HIF)-1α by inhibiting HIF-1α prolyl hydroxylation, which promotes glycolytic energy metabolism, vasculogenesis and angiogenesis, whereas HIF-1α is degraded by the HIF prolyl hydroxylase under normoxic conditions. We focus this review on the links between the activated Wnt/β-catenin pathway and the mechanisms underlying vasculogenesis and angiogenesis through HIF-1α under normoxic conditions in gliomas. Wnt-induced epidermal growth factor receptor/phosphatidylinositol 3-kinase (PI3K)/Akt signaling, Wnt-induced signal transducers and activators of transcription 3 (STAT3) signaling, and Wnt/β-catenin target gene transduction (c-Myc) can activate HIF-1α in a hypoxia-independent manner. The PI3K/Akt/mammalian target of rapamycin pathway activates HIF-1α through eukaryotic translation initiation factor 4E-binding protein 1 and STAT3. The β-catenin/T-cell factor 4 complex directly binds to STAT3 and activates HIF-1α, which up-regulates the Wnt/β-catenin target genes cyclin D1 and c-Myc in a positive feedback loop. Phosphorylated STAT3 by interleukin-6 or leukemia inhibitory factor activates HIF-1α even under normoxic conditions. The activation of the Wnt/β-catenin pathway induces, via the Wnt target genes c-Myc and cyclin D1 or via HIF-1α, gene transactivation encoding aerobic glycolysis enzymes, such as glucose transporter, hexokinase 2, pyruvate kinase M2, pyruvate dehydrogenase kinase 1 and lactate dehydrogenase-A, leading to lactate production, as the primary alternative of ATP, at all oxygen levels, even in normoxic conditions. Lactate released by glioma cells via the monocarboxylate lactate transporter-1 up-regulated by HIF-1α and lactate anion activates HIF-1α in normoxic endothelial cells by inhibiting HIF-1α prolyl hydroxylation and preventing HIF labeling by the von Hippel-Lindau protein. Increased lactate with acid environment and HIF-1α overexpression induce the vascular endothelial growth factor (VEGF) pathway of vasculogenesis and angiogenesis under normoxic conditions. Hypoxia and acidic pH have no synergistic effect on VEGF transcription.
Collapse
Affiliation(s)
- Alexandre Vallée
- Experimental and Clinical Neurosciences Laboratory, INSERM U1084, University of Poitiers, 11 Boulevard Marie et Pierre Curie, F-86000 Poitiers, France
| | - Rémy Guillevin
- DACTIM, UMR CNRS 7348, Université de Poitiers et CHU de Poitiers, F-86000 Poitiers, France
| | - Jean-Noël Vallée
- Laboratoire de Mathématiques et Applications (LMA), UMR CNRS 7348, University of Poitiers, F-86000 Poitiers, France
| |
Collapse
|
169
|
Zhang X, Chen J, Ai Z, Zhang Z, Lin L, Wei H. Targeting glycometabolic reprogramming to restore the sensitivity of leukemia drug-resistant K562/ADM cells to adriamycin. Life Sci 2018; 215:1-10. [PMID: 30473023 DOI: 10.1016/j.lfs.2018.10.050] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 10/16/2018] [Accepted: 10/24/2018] [Indexed: 02/04/2023]
Abstract
AIMS Mounting studies have confirmed that cancer cells reprogram their metabolism during early carcinogenesis to develop many other hallmarks, and demonstrated a relationship between aerobic glycolysis and the occurrence of drug resistance. However, the molecular mechanisms and role in tumor drug resistance of aerobic glycolysis remain unclear. MAIN METHODS We analyzed differentially expressed genes (DEGs) at the RNA level between the multi-drug resistance (MDR) leukemia cell line K562/adriamycin (ADM) and its parental, drug-sensitive K562 cell line. Clustering and enrichment analysis of DEGs was performed. Oxamate, a lactic dehydrogenase inhibitor were used to assess the effect of glycolysis inhibition on ADM susceptibility and the expression of the enriched DEGs in K562/ADM cells. KEY FINDINGS A total of 1742 DEGs were detected between the K562/ADM and K562 cell lines. The differential expression of unigenes encoding enzymes involved in glycometabolism signifies that there was a greater aerobic glycolysis flux in K562/ADM cells. The PI3K-AKT signaling pathway, which is related to glucose metabolism, showed representative differential enrichment and up-regulation in K562/ADM cells. Oxamate improved and re-sensitized the therapeutic effect of ADM in ADM-resistant cells by inhibiting aerobic glycolysis either directly or indirectly by down-regulation of the AKT-mTOR pathway. SIGNIFICANCE Our findings suggest that ADM resistance mediated by the increase of aerobic glycolysis, which related to the over-activation of the AKT-mTOR-c-Myc pathway in MDR leukemia cells. Inhibition of aerobic glycolysis and down-regulation of signaling pathways involved in aerobic glycolysis represent a potential chemotherapeutic strategy for sensitizing leukemic cells and thereby overcoming MDR.
Collapse
Affiliation(s)
- Xueyan Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Jing Chen
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Ziying Ai
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Zhewen Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Li Lin
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Hulai Wei
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China.
| |
Collapse
|
170
|
|
171
|
Marbaniang C, Kma L. Dysregulation of Glucose Metabolism by Oncogenes and Tumor Suppressors in Cancer Cells. Asian Pac J Cancer Prev 2018; 19:2377-2390. [PMID: 30255690 PMCID: PMC6249467 DOI: 10.22034/apjcp.2018.19.9.2377] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 08/20/2018] [Indexed: 02/07/2023] Open
Abstract
Cancers are complex diseases having several unique features, commonly described as ‘hallmarks of cancer’. Among them, altered signaling pathways are the common characteristic features that drive cancer progression; this is achieved due to mutations that lead to the activation of growth promoting(s) oncogenes and inactivation of tumor suppressors. As a result of which, cancer cells increase their glycolytic rate by consuming a large amount of glucose, and convert a majority of glucose to lactate even in the presence of oxygen known as the “Warburg effect”. Tumor cells like other cells are strictly dependent on energy for growth and survival; therefore, understanding energy metabolism will give us an idea to develop new effective anti-cancer therapies that target cancer energy production pathways. This review summarizes the roles of tumor suppressors and oncogenes and their products that provide metabolic advantages to cancer cells which in turn leads to the establishment of the “Warburg effect” and ultimately leads to cancer progression. Understanding cancer cell’s vulnerability will provide potential targets for its control.
Collapse
Affiliation(s)
- Casterland Marbaniang
- Department of Biochemistry, Cancer and Radiation Countermeasures Unit,North-Eastern Hill University, Shillong, Meghalaya, India.
| | | |
Collapse
|
172
|
Wang G, Wang JJ, Yin PH, Xu K, Wang YZ, Shi F, Gao J, Fu XL. New strategies for targeting glucose metabolism-mediated acidosis for colorectal cancer therapy. J Cell Physiol 2018; 234:348-368. [PMID: 30069931 DOI: 10.1002/jcp.26917] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 06/13/2018] [Indexed: 12/15/2022]
Abstract
Colorectal cancer (CRC) is a heterogeneous group of diseases that are the result of abnormal glucose metabolism alterations with high lactate production by pyruvate to lactate conversion, which remodels acidosis and offers an evolutional advantage for tumor cells, even enhancing their aggressive phenotype. This review summarizes recent findings that involve multiple genes, molecules, and downstream signaling in the dysregulated glycolytic pathway, which can allow a tumor to initiate acid byproducts and to progress, thereby resulting in acidosis commonly found in the tumor microenvironment of CRC. Moreover, the relationship between CRC cells and the tumor acidic microenvironment, especially for regulating lactate production and lactate dehydrogenase A levels, is also discussed, as well as comprehensively defining different aspects of glycolytic pathways that affect cancer cell proliferation, invasion, and migration. Furthermore, this review concentrates on glucose metabolism-mediated transduction factors in CRC, which include acid-sensing ion channels, triosephosphate isomerase and key glycolysis-related enzymes that regulate glycolytic metabolites, coupled with the effect on tumor cell glycolysis as well as signaling pathways. In conclusion, glucose metabolism mediated by glycolytic pathways that are integral to tumor acidosis in CRC is demonstrated. Therefore, selective metabolic inhibitors or agents against these targets in glucose metabolism through glycolytic pathways may be clinically useful to regulate the tumor's acidic microenvironment for CRC treatment and to identify specific targets that regulate tumor acidosis through a cancer patient-personalized approach. Furthermore, strategies for modifying the metabolic processes that effectively inhibit cancer cell growth and tumor progression and activate potent anticancer effects may provide more effective antitumor prospects for CRC therapy.
Collapse
Affiliation(s)
- Gang Wang
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| | - Jun-Jie Wang
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| | - Pei-Hao Yin
- Department of Cancer, Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ke Xu
- Department of Cancer, Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu-Zhu Wang
- Department of Medicine, Jiangsu University, Zhenjiang, China
| | - Feng Shi
- Department of Medicine, Jiangsu University, Zhenjiang, China
| | - Jing Gao
- Department of Medicine, Jiangsu University, Zhenjiang, China
| | - Xing-Li Fu
- Department of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
173
|
Do SK, Jeong JY, Lee SY, Choi JE, Hong MJ, Kang HG, Lee WK, Seok Y, Lee EB, Shin KM, Yoo SS, Lee J, Cha SI, Kim CH, Neugent ML, Goodwin J, Kim JW, Park JY. Glucose Transporter 1 Gene Variants Predict the Prognosis of Patients with Early-Stage Non-small Cell Lung Cancer. Ann Surg Oncol 2018; 25:3396-3403. [PMID: 30062472 DOI: 10.1245/s10434-018-6677-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Indexed: 12/27/2022]
Abstract
BACKGROUND This study was conducted to investigate whether polymorphisms of glucose transporter 1 (GLUT1) gene are associated with the prognosis of patients with non-small cell lung cancer (NSCLC) after surgical resection. METHODS Five single nucleotide polymorphisms (SNPs) in GLUT1 were investigated in a total of 354 patients with NSCLC who underwent curative surgery. The association of the SNPs with patients' survival was analyzed. RESULTS Among the five SNPs investigated, two SNPs (GLUT1 rs3820589T > A and rs4658G > C) were significantly associated with OS in multivariate analyses. GLUT1 rs3820589T > A was associated with significantly better OS (adjusted hazard ratio [aHR] = 0.57, 95% confidence interval [CI] = 0.34-0.94, P = 0.03, under dominant model), and rs4658G > C was associated with significantly worse OS (aHR = 1.91, 95% CI = 1.09-3.33, P = 0.02, under recessive model). In the stratified analysis by tumor histology, the effect of these SNPs on OS was only significant in squamous cell carcinoma but not in adenocarcinoma. When the two SNPs were combined, OS decreased as the number of bad genotypes increased (Ptrend = 4 × 10-3). CONCLUSIONS This study suggests that genetic variation in GLUT1 may be useful in predicting survival of patients with early stage NSCLC.
Collapse
Affiliation(s)
- Sook Kyung Do
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, Kyungpook National University, Daegu, Republic of Korea
| | - Ji Yun Jeong
- Department of Pathology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Shin Yup Lee
- Lung Cancer Center, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea. .,Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.
| | - Jin Eun Choi
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Mi Jeong Hong
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Hyo-Gyoung Kang
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Won Kee Lee
- Medical Research Collaboration Center in Kyungpook National University Hospital and School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Yangki Seok
- Lung Cancer Center, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea.,Department of Thoracic Surgery, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Eung Bae Lee
- Lung Cancer Center, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea.,Department of Thoracic Surgery, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Kyung Min Shin
- Department of Radiology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Seung Soo Yoo
- Lung Cancer Center, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea.,Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Jaehee Lee
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Seung Ick Cha
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Chang Ho Kim
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Michael L Neugent
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Justin Goodwin
- Yale School of Medicine, New Haven, CT, USA.,Yale Graduate School of Arts and Sciences, New Haven, CT, USA
| | - Jung-Whan Kim
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Jae Yong Park
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea. .,BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, Kyungpook National University, Daegu, Republic of Korea. .,Lung Cancer Center, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea. .,Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea. .,Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.
| |
Collapse
|
174
|
Blevins MA, Zhang C, Zhang L, Li H, Li X, Norris DA, Huang M, Zhao R. CPP-E1A fusion peptides inhibit CtBP-mediated transcriptional repression. Mol Oncol 2018; 12:1358-1373. [PMID: 29879296 PMCID: PMC6068344 DOI: 10.1002/1878-0261.12330] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 05/17/2018] [Accepted: 05/18/2018] [Indexed: 12/29/2022] Open
Abstract
The carboxyl‐terminal binding proteins (CtBP) are transcriptional corepressors that regulate the expression of multiple epithelial‐specific and pro‐apoptotic genes. Overexpression of CtBP occurs in many human cancers where they promote the epithelial‐to‐mesenchymal transition, stem cell‐like features, and cell survival, while knockdown of CtBP in tumor cells results in p53‐independent apoptosis. CtBPs are recruited to their target genes by binding to a conserved PXDLS peptide motif present in multiple DNA‐binding transcription factors. Disrupting the interaction between CtBP and its transcription factor partners may be a means of altering CtBP‐mediated transcriptional repression and a potential approach for cancer therapies. However, small molecules targeting protein–protein interactions have traditionally been difficult to identify. In this study, we took advantage of the fact that CtBP binds to a conserved peptide motif to explore the feasibility of using peptides containing the PXDLS motif fused to cell‐penetrating peptides (CPP) to inhibit CtBP function. We demonstrate that these peptides disrupt the ability of CtBP to interact with its protein partner, E1A, in an AlphaScreen assay. Moreover, these peptides can enter both lung carcinoma and melanoma cells, disrupt the interaction between CtBP and a transcription factor partner, and inhibit CtBP‐mediated transcriptional repression. Finally, the constitutive expression of one such peptide, Pep1‐E1A‐WT, in a melanoma cell line reverses CtBP‐mediated oncogenic phenotypes including proliferation, migration, and sphere formation and limits tumor growth in vivo. Together, our results suggest that CPP‐fused PXDLS‐containing peptides can potentially be developed into a research tool or therapeutic agent targeting CtBP‐mediated transcriptional events in various biological pathways.
Collapse
Affiliation(s)
- Melanie A Blevins
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Caiguo Zhang
- Department of Dermatology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Lingdi Zhang
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Hong Li
- Department of Dermatology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Xueni Li
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - David A Norris
- Department of Dermatology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Mingxia Huang
- Department of Dermatology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Rui Zhao
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
175
|
Lambert M, Jambon S, Depauw S, David-Cordonnier MH. Targeting Transcription Factors for Cancer Treatment. Molecules 2018; 23:molecules23061479. [PMID: 29921764 PMCID: PMC6100431 DOI: 10.3390/molecules23061479] [Citation(s) in RCA: 249] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 06/11/2018] [Accepted: 06/15/2018] [Indexed: 12/15/2022] Open
Abstract
Transcription factors are involved in a large number of human diseases such as cancers for which they account for about 20% of all oncogenes identified so far. For long time, with the exception of ligand-inducible nuclear receptors, transcription factors were considered as “undruggable” targets. Advances knowledge of these transcription factors, in terms of structure, function (expression, degradation, interaction with co-factors and other proteins) and the dynamics of their mode of binding to DNA has changed this postulate and paved the way for new therapies targeted against transcription factors. Here, we discuss various ways to target transcription factors in cancer models: by modulating their expression or degradation, by blocking protein/protein interactions, by targeting the transcription factor itself to prevent its DNA binding either through a binding pocket or at the DNA-interacting site, some of these inhibitors being currently used or evaluated for cancer treatment. Such different targeting of transcription factors by small molecules is facilitated by modern chemistry developing a wide variety of original molecules designed to specifically abort transcription factor and by an increased knowledge of their pathological implication through the use of new technologies in order to make it possible to improve therapeutic control of transcription factor oncogenic functions.
Collapse
Affiliation(s)
- Mélanie Lambert
- INSERM UMR-S1172-JPARC (Jean-Pierre Aubert Research Center), Lille University and Hospital Center (CHU-Lille), Institut pour la Recherche sur le Cancer de Lille (IRCL), Place de Verdun, F-59045 Lille, France.
| | - Samy Jambon
- INSERM UMR-S1172-JPARC (Jean-Pierre Aubert Research Center), Lille University and Hospital Center (CHU-Lille), Institut pour la Recherche sur le Cancer de Lille (IRCL), Place de Verdun, F-59045 Lille, France.
| | - Sabine Depauw
- INSERM UMR-S1172-JPARC (Jean-Pierre Aubert Research Center), Lille University and Hospital Center (CHU-Lille), Institut pour la Recherche sur le Cancer de Lille (IRCL), Place de Verdun, F-59045 Lille, France.
| | - Marie-Hélène David-Cordonnier
- INSERM UMR-S1172-JPARC (Jean-Pierre Aubert Research Center), Lille University and Hospital Center (CHU-Lille), Institut pour la Recherche sur le Cancer de Lille (IRCL), Place de Verdun, F-59045 Lille, France.
| |
Collapse
|
176
|
Metabolic Regulation in Mitochondria and Drug Resistance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1038:149-171. [PMID: 29178075 DOI: 10.1007/978-981-10-6674-0_11] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mitochondria are generally considered as a powerhouse in a cell where the majority of the cellular ATP and metabolite productions occur. Metabolic rewiring and reprogramming may be initiated and regulated by mitochondrial enzymes. The hypothesis that cellular metabolic rewiring and reprogramming processes may occur as cellular microenvironment is disturbed, resulting in alteration of cell phenotype, such as cancer cells resistant to therapeutics seems to be now acceptable. Cancer metabolic reprogramming regulated by mitochondrial enzymes is now one of the hallmarks of cancer. This chapter provides an overview of cancer metabolism and summarizes progress made in mitochondria-mediated metabolic regulation in cancer drug resistance.
Collapse
|
177
|
Piccaluga PP, Weber A, Ambrosio MR, Ahmed Y, Leoncini L. Epstein-Barr Virus-Induced Metabolic Rearrangements in Human B-Cell Lymphomas. Front Microbiol 2018; 9:1233. [PMID: 29937761 PMCID: PMC6002739 DOI: 10.3389/fmicb.2018.01233] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 05/22/2018] [Indexed: 12/16/2022] Open
Abstract
Tumor metabolism has been the object of several studies in the past, leading to the pivotal observation of a consistent shift toward aerobic glycolysis (so-called Warburg effect). More recently, several additional investigations proved that tumor metabolism is profoundly affected during tumorigenesis, including glucose, lipid and amino-acid metabolism. It is noticeable that metabolic reprogramming can represent a suitable therapeutic target in many cancer types. Epstein-Barr virus (EBV) was the first virus linked with cancer in humans when Burkitt lymphoma (BL) was described. Besides other well-known effects, it was recently demonstrated that EBV can induce significant modification in cell metabolism, which may lead or contribute to neoplastic transformation of human cells. Similarly, virus-induced tumorigenesis is characterized by relevant metabolic abnormalities directly induced by the oncoviruses. In this article, the authors critically review the most recent literature concerning EBV-induced metabolism alterations in lymphomas.
Collapse
Affiliation(s)
- Pier P. Piccaluga
- Department of Experimental, Diagnostic, and Specialty Medicine, Bologna University School of Medicine, Bologna, Italy
- Euro-Mediterranean Institute of Science and Technology, Palermo, Italy
- Department of Pathology, Jomo Kenyatta University of Agriculture and Technology, Juja, Kenya
| | - Alessandra Weber
- Department of Experimental, Diagnostic, and Specialty Medicine, Bologna University School of Medicine, Bologna, Italy
| | - Maria R. Ambrosio
- Section of Pathology, Department of Medical Biotechnology, University of Siena, Siena, Italy
| | - Yonis Ahmed
- Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Lorenzo Leoncini
- Section of Pathology, Department of Medical Biotechnology, University of Siena, Siena, Italy
| |
Collapse
|
178
|
Shima K, Kaeding N, Ogunsulire IM, Kaufhold I, Klinger M, Rupp J. Interferon-γ interferes with host cell metabolism during intracellular Chlamydia trachomatis infection. Cytokine 2018; 112:95-101. [PMID: 29885991 DOI: 10.1016/j.cyto.2018.05.039] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 05/18/2018] [Accepted: 05/31/2018] [Indexed: 12/11/2022]
Abstract
Interferon-γ (IFN-γ) is a central mediator of host immune responses including T-cell differentiation and activation of macrophages for the control of bacterial pathogens. Anti-bacterial mechanisms of IFN-γ against the obligate intracellular bacteria Chlamydiatrachomatis in epithelial cells have been intensively investigated in the past, focusing on cellular tryptophan depletion by an IFN-γ induced expression of the indoleamine 2, 3-deoxygenase (IDO). In this study, we could show that IFN-γ treatment caused a significant reduction of the host cell glycolysis that was accompanied by a reduction of glucose transporter-1 (GLUT1) and hypoxia inducible factor-1α (HIF-1α) expression. Furthermore, C. trachomatis induced enhancement of glycolytic and mitochondrial activation were significantly suppressed by IFN-γ treatment. We could further show that glucose starvation, as observed under IFN-γ treatment, was associated with an attenuated antimicrobial efficacy of doxycycline (DOX) against C. trachomatis. In conclusions, anti-chlamydial activity of IFN-γ goes beyond tryptophan depletion including interference with cellular energy metabolism resulting reduced progeny, but also impaired antimicrobial susceptibility of C. trachomatis.
Collapse
Affiliation(s)
- Kensuke Shima
- Department of Infectious Diseases and Microbiology, University of Luebeck, Luebeck, Germany.
| | - Nadja Kaeding
- Department of Infectious Diseases and Microbiology, University of Luebeck, Luebeck, Germany
| | | | - Inga Kaufhold
- Department of Infectious Diseases and Microbiology, University of Luebeck, Luebeck, Germany
| | | | - Jan Rupp
- Department of Infectious Diseases and Microbiology, University of Luebeck, Luebeck, Germany; German Center for Infection Research (DZIF), Partner Site Hamburg-Luebeck-Borstel, Germany
| |
Collapse
|
179
|
Ohana N, Benharroch D, Sheinis D, Cohen A. Traumatic glioblastoma: commentary and suggested mechanism. J Int Med Res 2018; 46:2170-2176. [PMID: 29708004 PMCID: PMC6023070 DOI: 10.1177/0300060518771265] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 03/26/2018] [Indexed: 12/17/2022] Open
Abstract
The role of head trauma in the development of glioblastoma is highly controversial and has been minimized since first put forward. This is not unexpected because skull injuries are overwhelmingly more common than glioblastoma. This paper presents a commentary based on the contributions of James Ewing, who established a major set of criteria for the recognition of an official relationship between trauma and cancer. Ewing's criteria were very stringent. The scholars who succeeded Ewing have facilitated the characterization of traumatic brain injuries since the introduction of computed tomography and magnetic resonance imaging. Discussions of the various criteria that have since developed are now being conducted, and those of an unnecessarily limiting nature are being highlighted. Three transcription factors associated with traumatic brain injury have been identified: p53, hypoxia-inducible factor-1α, and c-MYC. A role for these three transcription factors in the relationship between traumatic brain injury and glioblastoma is suggested; this role may support a cause-and-effect link with the subsequent development of glioblastoma.
Collapse
Affiliation(s)
- Nissim Ohana
- Surgical Orthopedics, Soroka University Medical Center and Faculty of Heath Sciences, Ben Gurion University of the Negev, Beer-Sheva, Israel
| | - Daniel Benharroch
- Department of Pathology, Soroka University Medical Center and Faculty of Health Sciences, Ben Gurion University of the Negev, Beer-Sheva, Israel
| | - Dimitri Sheinis
- Surgical Orthopedics, Soroka University Medical Center and Faculty of Heath Sciences, Ben Gurion University of the Negev, Beer-Sheva, Israel
| | - Abraham Cohen
- Neurosurgery Department, Soroka University Medical Center and Faculty of Health Sciences, Ben Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
180
|
Guo Q, Lan F, Yan X, Xiao Z, Wu Y, Zhang Q. Hypoxia exposure induced cisplatin resistance partially via activating p53 and hypoxia inducible factor-1α in non-small cell lung cancer A549 cells. Oncol Lett 2018; 16:801-808. [PMID: 29971135 PMCID: PMC6019907 DOI: 10.3892/ol.2018.8767] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 04/11/2018] [Indexed: 01/28/2023] Open
Abstract
Lung cancer is one of the most frequently occurring and fatal cancer types worldwide. Cisplatin is widely used for chemotherapy of non-small cell lung cancer (NSCLC). However, the use of cisplatin has been met with the challenge of chemoresistance as a result of hypoxia, which is common in adult solid tumors and is a principal cause of a poor patient outcome. In the present study, the effects of hypoxia on the response of the NSCLC A549 cell line to the clinically relevant cytotoxic cisplatin were evaluated via regulating hypoxia inducible facor-1α (HIF-1α) and p53. Hypoxia exposure upregulated the expression levels of HIF-1α and p53, and promoted glycolysis in A549 cells, which was attenuated by HIF-1α knockdown by siRNA introduction, indicating the critical roles of HIF-1α in regulating glycolysis under hypoxic conditions. HIF-1α-knockdown also sensitized A549 cells to cisplatin in hypoxia-exposed, but not in normoxia-exposed A549 cells, suggesting that hypoxia-induced cisplatin resistance partially contributes toward the upregulation of HIF-1α by hypoxia exposure. The present study also determined that hypoxia-upregulated p53 activated its downstream target gene p21 transcriptionally and blocked the cell cycle at the G1-G0 phase, thereby leading to inhibition of cell proliferation. As a result, activated p53 desensitized A549 cells to cisplatin potentially through increasing the non-proliferation status of A549 cells and therefore minimizing the influence of cisplatin. Taken together, these results identified the exact effects of HIF-1α and p53 induced by hypoxia and potentially elucidated their protective effects on A549 cells against cisplatin.
Collapse
Affiliation(s)
- Qiang Guo
- Department of Endocrinology, Chengdu First People's Hospital, Chengdu, Sichuan 610000, P.R. China
| | - Fei Lan
- Department of Endocrinology, Chengdu First People's Hospital, Chengdu, Sichuan 610000, P.R. China
| | - Xu Yan
- Department of Endocrinology, Chengdu First People's Hospital, Chengdu, Sichuan 610000, P.R. China
| | - Zhu Xiao
- Department of Endocrinology, Chengdu First People's Hospital, Chengdu, Sichuan 610000, P.R. China
| | - Yuelei Wu
- Department of Endocrinology, Chengdu First People's Hospital, Chengdu, Sichuan 610000, P.R. China
| | - Qin Zhang
- Department of Endocrinology, Chengdu First People's Hospital, Chengdu, Sichuan 610000, P.R. China
| |
Collapse
|
181
|
Schcolnik-Cabrera A, Chávez-Blanco A, Domínguez-Gómez G, Taja-Chayeb L, Morales-Barcenas R, Trejo-Becerril C, Perez-Cardenas E, Gonzalez-Fierro A, Dueñas-González A. Orlistat as a FASN inhibitor and multitargeted agent for cancer therapy. Expert Opin Investig Drugs 2018; 27:475-489. [PMID: 29723075 DOI: 10.1080/13543784.2018.1471132] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Cancer cells have increased glycolysis and glutaminolysis. Their third feature is increased de novo lipogenesis. As such, fatty acid (FA) synthesis enzymes are over-expressed in cancer and their depletion causes antitumor effects. As fatty acid synthase (FASN) plays a pivotal role in this process, it is an attractive target for cancer therapy. AREAS COVERED This is a review of the lipogenic phenotype of cancer and how this phenomenon can be exploited for cancer therapy using inhibitors of FASN, with particular emphasis on orlistat as a repurposing drug. EXPERT OPINION Disease stabilization only has been observed with a highly selective FASN inhibitor used as a single agent in clinical trials. It is too early to say whether the absence of tumor responses other than stabilization results because even full inhibition of FASN is not enough to elicit antitumor responses. The FASN inhibitor orlistat is a 'dirty' drug with target-off actions upon at least seven targets with a proven role in tumor biology. The development of orlistat formulations suited for its intravenous administration is a step ahead to shed light on the concept that drug promiscuity can or not be a virtue.
Collapse
Affiliation(s)
| | - Alma Chávez-Blanco
- a Division of Basic Research , Instituto Nacional de Cancerologia , Mexico City , Mexico
| | | | - Lucia Taja-Chayeb
- a Division of Basic Research , Instituto Nacional de Cancerologia , Mexico City , Mexico
| | - Rocio Morales-Barcenas
- a Division of Basic Research , Instituto Nacional de Cancerologia , Mexico City , Mexico
| | | | - Enrique Perez-Cardenas
- a Division of Basic Research , Instituto Nacional de Cancerologia , Mexico City , Mexico
| | - Aurora Gonzalez-Fierro
- a Division of Basic Research , Instituto Nacional de Cancerologia , Mexico City , Mexico
| | - Alfonso Dueñas-González
- b Unit of Biomedical Research in Cancer , Instituto de Investigaciones Biomedicas, UNAM/Instituto Nacional de Cancerologia , Mexico City , Mexico
| |
Collapse
|
182
|
Arif T, Krelin Y, Nakdimon I, Benharroch D, Paul A, Dadon-Klein D, Shoshan-Barmatz V. VDAC1 is a molecular target in glioblastoma, with its depletion leading to reprogrammed metabolism and reversed oncogenic properties. Neuro Oncol 2018; 19:951-964. [PMID: 28339833 DOI: 10.1093/neuonc/now297] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background Glioblastoma (GBM), an aggressive brain tumor with frequent relapses and a high mortality, still awaits an effective treatment. Like many cancers, GBM cells acquire oncogenic properties, including metabolic reprogramming, vital for growth. As such, tumor metabolism is an emerging avenue for cancer therapy. One relevant target is the voltage-dependent anion channel 1 (VDAC1), a mitochondrial protein controlling cell energy and metabolic homeostasis. Methods We used VDAC1-specific short interfering (si)RNA (si-VDAC1) to treat GBM cell lines and subcutaneous or intracranial-orthotopic GBM xenograft mouse models. Tumors were monitored using MRI, immunohistochemistry, immunoblotting, immunofluorescence, quantitative real-time PCR, transcription factor expression, and DNA microarray analyses. Results Silencing VDAC1 expression using si-VDAC1 in 9 glioblastoma-related cell lines, including patient-derived cells, led to marked decreases in VDAC1 levels and cell growth. Using si-VDAC1 in subcutaneous or intracranial-orthotopic GBM models inhibited tumor growth and reversed oncogenic properties, such as reprogrammed metabolism, stemness, angiogenesis, epithelial-mesenchymal transition, and invasiveness. In cells in culture, si-VDAC1 inhibits cancer neurosphere formation and, in tumors, targeted cancer stem cells, leading to their differentiation into neuronal-like cells. These VDAC1 depletion-mediated effects involved alterations in transcription factors regulating signaling pathways associated with cancer hallmarks. Conclusion VDAC1 offers a target for GBM treatment, allowing for attacks on the interplay between metabolism and oncogenic signaling networks, leading to tumor cell differentiation into neuron- and astrocyte-like cells. Simultaneously attacking all of these processes, VDAC1 depletion overcame GBM heterogeneity and can replace several anticancer drugs that separately target angiogenesis, proliferation, or metabolism.
Collapse
Affiliation(s)
- Tasleem Arif
- Department of Life Sciences, and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev,Beer-Sheva, Israel; Department of Pathology, Soroka University Medical Centre, and Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Yakov Krelin
- Department of Life Sciences, and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev,Beer-Sheva, Israel; Department of Pathology, Soroka University Medical Centre, and Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Itay Nakdimon
- Department of Life Sciences, and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev,Beer-Sheva, Israel; Department of Pathology, Soroka University Medical Centre, and Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Daniel Benharroch
- Department of Life Sciences, and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev,Beer-Sheva, Israel; Department of Pathology, Soroka University Medical Centre, and Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Avijit Paul
- Department of Life Sciences, and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev,Beer-Sheva, Israel; Department of Pathology, Soroka University Medical Centre, and Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Daniela Dadon-Klein
- Department of Life Sciences, and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev,Beer-Sheva, Israel; Department of Pathology, Soroka University Medical Centre, and Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Varda Shoshan-Barmatz
- Department of Life Sciences, and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev,Beer-Sheva, Israel; Department of Pathology, Soroka University Medical Centre, and Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
183
|
p53 and glucose metabolism: an orchestra to be directed in cancer therapy. Pharmacol Res 2018; 131:75-86. [DOI: 10.1016/j.phrs.2018.03.015] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 02/23/2018] [Accepted: 03/20/2018] [Indexed: 12/14/2022]
|
184
|
Anura A, Kazi A, Pal M, Paul RR, Sengupta S, Chatterjee J. Endorsing cellular competitiveness in aberrant epithelium of oral submucous fibrosis progression: neighbourhood analysis of immunohistochemical attributes. Histochem Cell Biol 2018; 150:61-75. [DOI: 10.1007/s00418-018-1671-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2018] [Indexed: 12/14/2022]
|
185
|
Londhe P, Yu PY, Ijiri Y, Ladner KJ, Fenger JM, London C, Houghton PJ, Guttridge DC. Classical NF-κB Metabolically Reprograms Sarcoma Cells Through Regulation of Hexokinase 2. Front Oncol 2018; 8:104. [PMID: 29696133 PMCID: PMC5904193 DOI: 10.3389/fonc.2018.00104] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 03/23/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Metabolic reprogramming has emerged as a cancer hallmark, and one of the well-known cancer-associated metabolic alterations is the increase in the rate of glycolysis. Recent reports have shown that both the classical and alternative signaling pathways of nuclear factor κB (NF-κB) play important roles in controlling the metabolic profiles of normal cells and cancer cells. However, how these signaling pathways affect the metabolism of sarcomas, specifically rhabdomyosarcoma (RMS) and osteosarcoma (OS), has not been characterized. METHODS Classical NF-κB activity was inhibited through overexpression of the IκBα super repressor of NF-κB in RMS and OS cells. Global gene expression analysis was performed using Affymetrix GeneChip Human Transcriptome Array 2.0, and data were interpreted using gene set enrichment analysis. Seahorse Bioscience XFe24 was used to analyze oxygen consumption rate as a measure of aerobic respiration. RESULTS Inhibition of classical NF-κB activity in sarcoma cell lines restored alternative signaling as well as an increased oxidative respiratory metabolic phenotype in vitro. In addition, microarray analysis indicated that inhibition of NF-κB in sarcoma cells reduced glycolysis. We showed that a glycolytic gene, hexokinase (HK) 2, is a direct NF-κB transcriptional target. Knockdown of HK2 shifted the metabolic profile in sarcoma cells away from aerobic glycolysis, and re-expression of HK2 rescued the metabolic shift induced by inhibition of NF-κB activity in OS cells. CONCLUSION These findings suggest that classical signaling of NF-κB plays a crucial role in the metabolic profile of pediatric sarcomas potentially through the regulation of HK2.
Collapse
Affiliation(s)
- Priya Londhe
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, United States
- Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Peter Y. Yu
- Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
- Medical Student Research Program, The Ohio State University, Columbus, OH, United States
| | - Yuichi Ijiri
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, United States
- Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Katherine J. Ladner
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, United States
- Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Joelle M. Fenger
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States
| | - Cheryl London
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States
- Cummings School of Veterinary Medicine, Tufts University, Grafton, MA, United States
| | - Peter J. Houghton
- Greehey Children’s Research Institute, University of Texas Health Science Center, San Antonio, TX, United States
| | - Denis C. Guttridge
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, United States
- Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
186
|
DNA damage response upon environmental contaminants: An exhausting work for genomic integrity. CURRENT OPINION IN TOXICOLOGY 2018. [DOI: 10.1016/j.cotox.2017.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
187
|
Blevins MA, Huang M, Zhao R. The Role of CtBP1 in Oncogenic Processes and Its Potential as a Therapeutic Target. Mol Cancer Ther 2018; 16:981-990. [PMID: 28576945 DOI: 10.1158/1535-7163.mct-16-0592] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 11/11/2016] [Accepted: 02/22/2017] [Indexed: 12/24/2022]
Abstract
Transcriptional corepressor proteins have emerged as an important facet of cancer etiology. These corepressor proteins are often altered by loss- or gain-of-function mutations, leading to transcriptional imbalance. Thus, research directed at expanding our current understanding of transcriptional corepressors could impact the future development of new cancer diagnostics, prognostics, and therapies. In this review, our current understanding of the CtBP corepressors, and their role in both development and disease, is discussed in detail. Importantly, the role of CtBP1 overexpression in adult tissues in promoting the progression of multiple cancer types through their ability to modulate the transcription of developmental genes ectopically is explored. CtBP1 overexpression is known to be protumorigenic and affects the regulation of gene networks associated with "cancer hallmarks" and malignant behavior, including increased cell survival, proliferation, migration, invasion, and the epithelial-mesenchymal transition. As a transcriptional regulator of broad developmental processes capable of promoting malignant growth in adult tissues, therapeutically targeting the CtBP1 corepressor has the potential to be an effective method for the treatment of diverse tumor types. Although efforts to develop CtBP1 inhibitors are still in the early stages, the current progress and the future perspectives of therapeutically targeting this transcriptional corepressor are also discussed. Mol Cancer Ther; 16(6); 981-90. ©2017 AACR.
Collapse
Affiliation(s)
- Melanie A Blevins
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado
| | - Mingxia Huang
- Department of Dermatology, University of Colorado School of Medicine, Aurora, Colorado.
| | - Rui Zhao
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado.
| |
Collapse
|
188
|
Huang L, Xu H, Peng G. TLR-mediated metabolic reprogramming in the tumor microenvironment: potential novel strategies for cancer immunotherapy. Cell Mol Immunol 2018; 15:428-437. [PMID: 29553135 PMCID: PMC6068099 DOI: 10.1038/cmi.2018.4] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 12/28/2017] [Accepted: 12/29/2017] [Indexed: 02/06/2023] Open
Abstract
Cellular energy metabolism not only promotes tumor cell growth and metastasis but also directs immune cell survival, proliferation and the ability to perform specific and functional immune responses within the tumor microenvironment. A better understanding of the molecular regulation of metabolism in different cell components in the tumor-suppressive microenvironment is critical for the development of effective strategies for human cancer treatments. Toll-like receptors (TLRs) have recently been recognized as critical factors involved in tumor pathogenesis, regulating both tumor cells and tumor-infiltrating innate and adaptive immune cells. However, little is known about the molecular crosstalk between TLR signaling and tumor or/and immune cell metabolism, although there is abundant expression of TLRs in these cells. In this review, we explore the functional role of TLR signaling in reprogramming cell metabolism in the tumor microenvironment. In particular, we discuss how malignant tumors regulate metabolism to support their growth and survival, summarize more recently identified metabolic profiles of different immune cell subsets and TLR-mediated regulation of cellular metabolism in both tumor and immune cells, and further explore potential strategies targeting cell metabolism for TLR-based cancer therapy. An improved understanding of these issues should open new avenues for the development of novel strategies via TLR-mediated metabolic reprogramming of the tumor microenvironment for cancer immunotherapy.
Collapse
Affiliation(s)
- Lan Huang
- Division of Infectious Diseases, Allergy and Immunology, Department of Internal Medicine, Saint Louis University School of Medicine, 63104, Saint Louis, MO, USA.,Department of Microbiology and Immunology, Jiangsu University School of Medicine, 212013, Zhenjiang, China
| | - Huaxi Xu
- Department of Microbiology and Immunology, Jiangsu University School of Medicine, 212013, Zhenjiang, China
| | - Guangyong Peng
- Division of Infectious Diseases, Allergy and Immunology, Department of Internal Medicine, Saint Louis University School of Medicine, 63104, Saint Louis, MO, USA.
| |
Collapse
|
189
|
Minami K, Taniguchi K, Sugito N, Kuranaga Y, Inamoto T, Takahara K, Takai T, Yoshikawa Y, Kiyama S, Akao Y, Azuma H. MiR-145 negatively regulates Warburg effect by silencing KLF4 and PTBP1 in bladder cancer cells. Oncotarget 2018; 8:33064-33077. [PMID: 28380435 PMCID: PMC5464850 DOI: 10.18632/oncotarget.16524] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 03/15/2017] [Indexed: 11/29/2022] Open
Abstract
The Warburg effect is a well-known feature in cancer-specific metabolism. We previously reported on the role of microRNA (miR)-145 as a tumor-suppressor in human bladder cancer (BC) cells. In this study, we reveal that miR-145 decreases the Warburg effect by silencing KLF4 in BC cells. The expression levels of miR-145 were significantly lower in clinical BC samples and BC cell lines compared to those in normal tissues and HUC cells. Luciferase assay results showed that miR-145 directly bound to 3′UTR of KLF4, which was shown to be overexpressed in the clinical BC samples using Western blot analysis and immunohistochemistry. Remarkable growth inhibition and apoptosis were induced by the ectopic expression of miR-145 or by the gene silencing of KLF4 (siR-KLF4). Also, Warburg effect-related genes such as PTBP1/PKMs were regulated by the transfection of BC cells with miR-145 or siR-KLF4. These results thus indicate that the miR-145/KLF4/PTBP1/PKMs axis is one of the critical pathways that maintain the Warburg effect in BC carcinogenesis. MiR-145 perturbed the Warburg effect by suppressing the KLF4/PTBP1/PKMs pathway in BC cells, resulting in significant cell growth inhibition.
Collapse
Affiliation(s)
- Koichiro Minami
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu 501-1193, Japan.,Department of Urology, Osaka Medical College, Takatsuki, Osaka 569-8686, Japan
| | - Kohei Taniguchi
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu 501-1193, Japan.,Department of General and Gastroenterological Surgery, Osaka Medical College, Takatsuki, Osaka 569-8686, Japan
| | - Nobuhiko Sugito
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu 501-1193, Japan
| | - Yuki Kuranaga
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu 501-1193, Japan
| | - Teruo Inamoto
- Department of Urology, Osaka Medical College, Takatsuki, Osaka 569-8686, Japan
| | - Kiyoshi Takahara
- Department of Urology, Osaka Medical College, Takatsuki, Osaka 569-8686, Japan
| | - Tomoaki Takai
- Department of Urology, Osaka Medical College, Takatsuki, Osaka 569-8686, Japan
| | - Yuki Yoshikawa
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu 501-1193, Japan.,Department of Urology, Osaka Medical College, Takatsuki, Osaka 569-8686, Japan
| | - Satoshi Kiyama
- Department of Urology, Osaka Medical College, Takatsuki, Osaka 569-8686, Japan
| | - Yukihiro Akao
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu 501-1193, Japan
| | - Haruhito Azuma
- Department of Urology, Osaka Medical College, Takatsuki, Osaka 569-8686, Japan
| |
Collapse
|
190
|
Li L, Liang Y, Kang L, Liu Y, Gao S, Chen S, Li Y, You W, Dong Q, Hong T, Yan Z, Jin S, Wang T, Zhao W, Mai H, Huang J, Han X, Ji Q, Song Q, Yang C, Zhao S, Xu X, Ye Q. Transcriptional Regulation of the Warburg Effect in Cancer by SIX1. Cancer Cell 2018; 33:368-385.e7. [PMID: 29455928 DOI: 10.1016/j.ccell.2018.01.010] [Citation(s) in RCA: 287] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 10/26/2017] [Accepted: 01/18/2018] [Indexed: 12/12/2022]
Abstract
Aerobic glycolysis (the Warburg effect) facilitates tumor growth, and drugs targeting aerobic glycolysis are being developed. However, how the Warburg effect is directly regulated is largely unknown. Here we show that transcription factor SIX1 directly increases the expression of many glycolytic genes, promoting the Warburg effect and tumor growth in vitro and in vivo. SIX1 regulates glycolysis through HBO1 and AIB1 histone acetyltransferases. Cancer-related SIX1 mutation increases its ability to promote aerobic glycolysis and tumor growth. SIX1 glycolytic function is directly repressed by microRNA-548a-3p, which is downregulated, inversely correlates with SIX1, and is a good predictor of prognosis in breast cancer patients. Thus, the microRNA-548a-3p/SIX1 axis strongly links aerobic glycolysis to carcinogenesis and may become a promising cancer therapeutic target.
Collapse
Affiliation(s)
- Ling Li
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing 100850, China
| | - Yingchun Liang
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing 100850, China
| | - Lei Kang
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing 100850, China; Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| | - Yang Liu
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing 100850, China; Department of Thoracic Surgery, PLA General Hospital, Beijing 100853, China
| | - Shan Gao
- CAS Key Laboratory of Biomedical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Siyu Chen
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing 100850, China; Department of Thoracic Surgery, PLA General Hospital, Beijing 100853, China
| | - Ying Li
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing 100850, China; Department of Oncology, PLA General Hospital, Beijing 100853, China
| | - Wenye You
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing 100850, China; Department of Oncology, PLA General Hospital, Beijing 100853, China
| | - Qian Dong
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing 100850, China
| | - Tian Hong
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing 100850, China
| | - Zhifeng Yan
- Department of Gynecology and Obstetrics, PLA General Hospital, Beijing 100853, China
| | - Shuai Jin
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing 100850, China; Department of Thoracic Surgery, PLA General Hospital, Beijing 100853, China
| | - Tao Wang
- Department of Oncology, 307 Hospital of People's Liberation Army, Beijing 100071, China
| | - Wei Zhao
- Department of Oncology, General Hospital of the PLA Rocket Force, Beijing 100088, China
| | - Haixing Mai
- Department of Urology, 307 Hospital of People's Liberation Army, Beijing 100071, China
| | - Jun Huang
- Department of Urology, 307 Hospital of People's Liberation Army, Beijing 100071, China
| | - Xiao Han
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing 100850, China
| | - Quanbo Ji
- Department of Orthopedics, PLA General Hospital, Beijing 100853, China
| | - Qi Song
- Department of Oncology, PLA General Hospital, Beijing 100853, China
| | - Chao Yang
- Department of Oncology, General Hospital of the PLA Rocket Force, Beijing 100088, China
| | - Shixin Zhao
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing 100850, China
| | - Xiaojie Xu
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing 100850, China.
| | - Qinong Ye
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing 100850, China.
| |
Collapse
|
191
|
Benke S, Agerer B, Haas L, Stöger M, Lercher A, Gabler L, Kiss I, Scinicariello S, Berger W, Bergthaler A, Obenauf AC, Versteeg GA. Human tripartite motif protein 52 is required for cell context-dependent proliferation. Oncotarget 2018; 9:13565-13581. [PMID: 29568378 PMCID: PMC5862599 DOI: 10.18632/oncotarget.24422] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 01/30/2018] [Indexed: 12/16/2022] Open
Abstract
Tripartite motif (TRIM) proteins have been shown to play important roles in cancer development and progression by modulating cell proliferation or resistance from cell death during non-homeostatic stress conditions found in tumor micro-environments. In this study, we set out to investigate the importance for cellular fitness of the virtually uncharacterized family member TRIM52. The human TRIM52 gene has arisen recently in evolution, making it unlikely that TRIM52 is required for basic cellular functions in normal cells. However, a recent genome-wide ablation screening study has suggested that TRIM52 may be essential for optimal proliferation or survival in certain genetic cancer backgrounds. Identifying genes which fit this concept of genetic context-dependent fitness in cancer cells is of interest as they are promising targets for tumor-specific therapy. We report here that TRIM52 ablation significantly diminished the proliferation of specific glioblastoma cell lines in cell culture and mouse xenografts by compromising their cell cycle progression in a p53-dependent manner. Together, our findings point to a non-redundant TRIM52 function that is required for optimal proliferation.
Collapse
Affiliation(s)
- Stefan Benke
- Department of Microbiology, Immunobiology, and Genetics, Max F. Perutz Laboratories, University of Vienna, Vienna Biocenter, Vienna 1030, Austria
| | - Benedikt Agerer
- Department of Microbiology, Immunobiology, and Genetics, Max F. Perutz Laboratories, University of Vienna, Vienna Biocenter, Vienna 1030, Austria
| | - Lisa Haas
- Research Institute of Molecular Pathology, Vienna Biocenter, Vienna 1030, Austria
| | - Martin Stöger
- Department of Microbiology, Immunobiology, and Genetics, Max F. Perutz Laboratories, University of Vienna, Vienna Biocenter, Vienna 1030, Austria
| | - Alexander Lercher
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria
| | - Lisa Gabler
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Vienna A-1090, Austria
| | - Izabella Kiss
- Department of Microbiology, Immunobiology, and Genetics, Max F. Perutz Laboratories, University of Vienna, Vienna Biocenter, Vienna 1030, Austria
| | - Sara Scinicariello
- Department of Microbiology, Immunobiology, and Genetics, Max F. Perutz Laboratories, University of Vienna, Vienna Biocenter, Vienna 1030, Austria
| | - Walter Berger
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Vienna A-1090, Austria
| | - Andreas Bergthaler
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria
| | - Anna C. Obenauf
- Research Institute of Molecular Pathology, Vienna Biocenter, Vienna 1030, Austria
| | - Gijs A. Versteeg
- Department of Microbiology, Immunobiology, and Genetics, Max F. Perutz Laboratories, University of Vienna, Vienna Biocenter, Vienna 1030, Austria
| |
Collapse
|
192
|
Jochmanova I, Pacak K. Pheochromocytoma: The First Metabolic Endocrine Cancer. Clin Cancer Res 2018; 22:5001-5011. [PMID: 27742786 DOI: 10.1158/1078-0432.ccr-16-0606] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 08/23/2016] [Indexed: 01/21/2023]
Abstract
Dysregulated metabolism is one of the key characteristics of cancer cells. The most prominent alterations are present during regulation of cell respiration, which leads to a switch from oxidative phosphorylation to aerobic glycolysis. This metabolic shift results in activation of numerous signaling and metabolic pathways supporting cell proliferation and survival. Recent progress in genetics and metabolomics has allowed us to take a closer look at the metabolic changes present in pheochromocytomas (PHEO) and paragangliomas (PGL). These neuroendocrine tumors often exhibit dysregulation of mitochondrial metabolism, which is driven by mutations in genes encoding Krebs cycle enzymes or by activation of hypoxia signaling. Present metabolic changes are involved in processes associated with tumorigenesis, invasiveness, metastasis, and resistance to various cancer therapies. In this review, we discuss the metabolic nature of PHEOs/PGLs and how unveiling the metabolic disturbances present in tumors could lead to identification of new biomarkers and personalized cancer therapies. Clin Cancer Res; 22(20); 5001-11. ©2016 AACR SEE ALL ARTICLES IN THIS CCR FOCUS SECTION, "ENDOCRINE CANCERS REVISING PARADIGMS".
Collapse
Affiliation(s)
- Ivana Jochmanova
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland. First Department of Internal Medicine, Medical Faculty of P.J. Šafárik University in Košice, Košice, Slovakia
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland.
| |
Collapse
|
193
|
LncRNA IDH1-AS1 links the functions of c-Myc and HIF1α via IDH1 to regulate the Warburg effect. Proc Natl Acad Sci U S A 2018; 115:E1465-E1474. [PMID: 29378948 PMCID: PMC5816142 DOI: 10.1073/pnas.1711257115] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
We report in this article that c-Myc-mediated repression of lncRNA IDH1-AS1 sustains activation of the Warburg effect by HIF1α under normoxic conditions. IDH1-AS1 would otherwise enhance IDH1 enzymatic activity through promoting its homodimerization, leading to increased production of α-KG, which, along with decreases in ROS levels similarly resulting from increased IDH1 activity, causes down-regulation of HIF1a and a reduction in glycolysis. Collectively, our results have identified a signaling axis c-Myc-(IDH1-AS1)-IDH1-αKG/ROS-HIF1α that is important for activation of the Warburg effect under normoxia. Moreover, the results reveal IDH1 as a member of c-Myc-responsive metabolic enzymes and demonstrate that c-Myc plays an important part in balancing mitochondrial respiration and glycolysis to ensure glycolysis be executed efficiently in cancer cells under normoxia. The oncoprotein c-Myc plays an important role in regulating glycolysis under normoxia; yet, in cancer cells, HIF1α, which is essential for driving glycolysis under hypoxia, is often up-regulated even in the presence of oxygen. The relationship between these two major regulators of the Warburg effect remains to be fully defined. Here we demonstrate that regulation of a long noncoding RNA (lncRNA), named IDH1-AS1, enables c-Myc to collaborate with HIF1α in activating the Warburg effect under normoxia. c-Myc transcriptionally repressed IDH1-AS1, which, upon expression, promoted homodimerization of IDH1 and thus enhanced its enzymatic activity. This resulted in increased α-KG and decreased ROS production and subsequent HIF1α down-regulation, leading to attenuation of glycolysis. Hence, c-Myc repression of IDH1-AS1 promotes activation of the Warburg effect by HIF1α. As such, IDH1-AS1 overexpression inhibited cell proliferation, whereas silencing of IDH1-AS1 promoted cell proliferation and cancer xenograft growth. Restoring IDH1-AS1 expression may therefore represent a potential metabolic approach for cancer treatment.
Collapse
|
194
|
Guimarães TA, Farias LC, Santos ES, de Carvalho Fraga CA, Orsini LA, de Freitas Teles L, Feltenberger JD, de Jesus SF, de Souza MG, Santos SHS, de Paula AMB, Gomez RS, Guimarães ALS. Metformin increases PDH and suppresses HIF-1α under hypoxic conditions and induces cell death in oral squamous cell carcinoma. Oncotarget 2018; 7:55057-55068. [PMID: 27474170 PMCID: PMC5342401 DOI: 10.18632/oncotarget.10842] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 07/09/2016] [Indexed: 01/18/2023] Open
Abstract
Background Metformin is a biguanide, belonging to the oral hypoglycemic agents and is a widely used in the treatment of type 2 diabetes. Evidence indicate that Metformin inhibits cell proliferation in several human cancers and inhibits the Warburg phenomenon in tumor cells. Results Low PDH levels were observed in OSCC, and Metformin promotes an increase in PDH levels in hypoxic conditions. Metformin also reduced HIF-1α mRNA and protein levels. Metformin demonstrated antiproliferative effects, inhibited migration, increased the number of apoptotic cells and increased the transcription of caspase 3. Objective The present study aims to explore the effects of Metformin in hypoxic conditions. Specifically, we focused on pyruvate dehydrogenase (PDH), (hypoxia-inducible factor 1α) HIF-1α levels and the oral squamous cell carcinoma (OSCC) cell phenotype. Additionally, we also investigated a theoretical consequence of Metformin treatment. Methods PDH levels in patients with OSCC and oral dysplasia were evaluated. Metformin was administered in vitro to test the effect of Metformin under hypoxic conditions. The results were complemented by Bioinformatics analyses. Conclusions In conclusion, our current findings show that Metformin reduces HIF-1α gene expression and increases PDH expression. Metformin inhibits cell proliferation and migration in the OSCC cell line model. Additionally, Metformin enhances the number of apoptotic cells and caspase 3 levels. Interestingly enough, Metformin did not increase the mutant p53 levels under hypoxic conditions.
Collapse
Affiliation(s)
- Talita Antunes Guimarães
- Department of Dentistry, Universidade Estadual de Montes Claros, Montes Claros, Minas Gerais, Brazil
| | - Lucyana Conceição Farias
- Department of Dentistry, Universidade Estadual de Montes Claros, Montes Claros, Minas Gerais, Brazil
| | - Eliane Sobrinho Santos
- Department of Dentistry, Universidade Estadual de Montes Claros, Montes Claros, Minas Gerais, Brazil.,Instituto Federal de Educação, Ciência e Tecnologia do Norte de Minas Gerais (IFNMG), Araçuaí, Minas Gerais, Brazil
| | - Carlos Alberto de Carvalho Fraga
- Faculdades Integradas Pitágoras, Montes Claros, Minas Gerais, Brazil.,Faculdades Unidas do Norte de Minas, Montes Claros, Minas Gerais, Brazil
| | - Lissur Azevedo Orsini
- Department of Clinical, Surgery and Oral Pathology, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Leandro de Freitas Teles
- Department of Dentistry, Universidade Estadual de Montes Claros, Montes Claros, Minas Gerais, Brazil
| | | | - Sabrin Ferreira de Jesus
- Department of Dentistry, Universidade Estadual de Montes Claros, Montes Claros, Minas Gerais, Brazil
| | | | - Sérgio Henrique Sousa Santos
- Institute of Agricultural Sciences, Food Engineering College, Universidade Federal de Minas Gerais (UFMG), Montes Claros, Minas Gerais, Brazil
| | | | - Ricardo Santiago Gomez
- Department of Clinical, Surgery and Oral Pathology, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - André Luiz Sena Guimarães
- Department of Dentistry, Universidade Estadual de Montes Claros, Montes Claros, Minas Gerais, Brazil
| |
Collapse
|
195
|
Feng J, Zhang Q, Zhou Y, Yu S, Hong L, Zhao S, Yang J, Wan H, Xu G, Zhang Y, Li C. Integration of Proteomics and Metabolomics Revealed Metabolite-Protein Networks in ACTH-Secreting Pituitary Adenoma. Front Endocrinol (Lausanne) 2018; 9:678. [PMID: 30532734 PMCID: PMC6266547 DOI: 10.3389/fendo.2018.00678] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 10/29/2018] [Indexed: 12/20/2022] Open
Abstract
An effective treatment for the management of adrenocorticotropic hormone-secreting pituitary adenomas (ACTH-PA) is currently lacking, although surgery is a treatment option. We have integrated information obtained at the metabolomic and proteomic levels to identify critical networks and signaling pathways that may play important roles in the metabolic regulation of ACTH-PA and therefore hopefully represent potential therapeutic targets. Six ACTH-PAs and seven normal pituitary glands were investigated via gas chromatography-mass spectrometry (GC-MS) analysis for metabolomics. Five ACTH-PAs and five normal pituitary glands were subjected to proteomics analysis via nano liquid chromatography tandem-mass spectrometry (nanoLC-MS/MS). The joint pathway analysis and network analysis was performed using MetaboAnalyst 3.0. software. There were significant differences of metabolites and protein expression levels between the ACTH-PAs and normal pituitary glands. A proteomic analysis identified 417 differentially expressed proteins that were significantly enriched in the Myc signaling pathway. The protein-metabolite joint pathway analysis showed that differentially expressed proteins and metabolites were significantly enriched in glycolysis/gluconeogenesis, pyruvate metabolism, citrate cycle (TCA cycle), and the fatty acid metabolism pathway in ACTH-PA. The protein-metabolite molecular interaction network identified from the metabolomics and proteomics investigation resulted in four subnetworks. Ten nodes in subnetwork 1 were the most significantly enriched in cell amino acid metabolism and pyrimidine nucleotide metabolism. Additionally, the metabolite-gene-disease interaction network established nine subnetworks. Ninety-two nodes in subnetwork 1 were the most significantly enriched in carboxylic acid metabolism and organic acid metabolism. The present study clarified the pathway networks that function in ACTH-PA. Our results demonstrated the presence of downregulated glycolysis and fatty acid synthesis in this tumor type. We also revealed that the Myc signaling pathway significantly participated in the metabolic changes and tumorigenesis of ACTH-PA. This data may provide biomarkers for ACTH-PA diagnosis and monitoring, and could also lead to the development of novel strategies for treating pituitary adenomas.
Collapse
Affiliation(s)
- Jie Feng
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Beijing Institute for Brain Disorders, Brain Tumor Center, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Qi Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yang Zhou
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Shenyuan Yu
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Lichuan Hong
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Sida Zhao
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jingjing Yang
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Hong Wan
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Guowang Xu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Yazhuo Zhang
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Beijing Institute for Brain Disorders, Brain Tumor Center, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Chuzhong Li
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Beijing Institute for Brain Disorders, Brain Tumor Center, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- *Correspondence: Chuzhong Li
| |
Collapse
|
196
|
Wang W, Cai Q, Zhou F, Liu J, Jin X, Ni P, Lu M, Wang G, Zhang J. Impaired pentose phosphate pathway in the development of 3D MCF-7 cells mediated intracellular redox disturbance and multi-cellular resistance without drug induction. Redox Biol 2017; 15:253-265. [PMID: 29291545 PMCID: PMC5752090 DOI: 10.1016/j.redox.2017.12.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 12/06/2017] [Accepted: 12/20/2017] [Indexed: 02/06/2023] Open
Abstract
Although metabolic reprogramming and redox imbalance are widely reported to be involved in chemo-resistance in cancer treatment, much more attention was paid to anti-cancer drug induced effect. Our previous studies showed that cancer cells can develop P-gp overexpression-mediated intrinsic drug resistance in the formation of 3D MCF-7 multi-cellular layers (MCLs) without any drug induction. However, whether metabolic reprogramming and redox imbalance functioned during this progress remained unrevealed. In our present study, LC-Q/TOF-MS and GC-MS were used in combination for analysing intracellular metabolites. The contribution of pentose phosphate pathway (PPP) and its related redox status were checked by chemical interfering and silencing/over-expression of glucose-6-phosphate dehydrogenase (G6PD). The downstream products of G6PD were assayed by quantitative real-time PCR, western blot and flow cytometry. Results showed that not only G6PD expression but also G6PD activity was significantly lowered along with 3D MCF-7 cells culture time. Impaired PPP disturbed redox-cycling, generated reactive oxygen species (ROS), which triggered cell cycle arrest and caused the switch to Chk2/p53/NF-κB pathway-mediated P-gp induction. Our results provided a new attempt to associate intrinsic small molecule metabolites (impaired PPP) communicating with cell signalling pathways through disturbed intracellular redox status to elucidate multi-cellular resistance (MCR) in 3D MCF-7 cells, which improved the understanding of the mechanisms of P-gp up-regulation in MCR with metabolomic and related redox status support. Impaired pentose phosphate pathway (PPP) in the development of 3D MCF-7 cells. PPP mediated intracellular redox disturbance and multi-cellular resistance (MCR). P-gp up-regulation in MCR with metabolomics and related redox status support. Intrinsic small molecule metabolites communicating with cell signalling pathway.
Collapse
Affiliation(s)
- Wenjie Wang
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, Jiangsu, China
| | - Qingyun Cai
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, Jiangsu, China
| | - Fang Zhou
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, Jiangsu, China
| | - Jiali Liu
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, Jiangsu, China
| | - Xiaoliang Jin
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, Jiangsu, China
| | - Ping Ni
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, Jiangsu, China
| | - Meng Lu
- Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China.
| | - Guangji Wang
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, Jiangsu, China.
| | - Jingwei Zhang
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, Jiangsu, China.
| |
Collapse
|
197
|
Simulation of Cellular Energy Restriction in Quiescence (ERiQ)-A Theoretical Model for Aging. BIOLOGY 2017; 6:biology6040044. [PMID: 29231906 PMCID: PMC5745449 DOI: 10.3390/biology6040044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 12/08/2017] [Accepted: 12/09/2017] [Indexed: 02/07/2023]
Abstract
Cellular responses to energy stress involve activation of pro-survival signaling nodes, compensation in regulatory pathways and adaptations in organelle function. Specifically, energy restriction in quiescent cells (ERiQ) through energetic perturbations causes adaptive changes in response to reduced ATP, NAD+ and NADP levels in a regulatory network spanned by AKT, NF-κB, p53 and mTOR. Based on the experimental ERiQ platform, we have constructed a minimalistic theoretical model consisting of feedback motifs that enable investigation of stress-signaling pathways. The computer simulations reveal responses to acute energetic perturbations, promoting cellular survival and recovery to homeostasis. We speculated that the very same stress mechanisms are activated during aging in post-mitotic cells. To test this hypothesis, we modified the model to be deficient in protein damage clearance and demonstrate the formation of energy stress. Contrasting the network’s pro-survival role in acute energetic challenges, conflicting responses in aging disrupt mitochondrial maintenance and contribute to a lockstep progression of decline when chronically activated. The model was analyzed by a local sensitivity analysis with respect to lifespan and makes predictions consistent with inhibitory and gain-of-function experiments in aging.
Collapse
|
198
|
Vallée A, Lecarpentier Y, Vallée JN. Thermodynamic Aspects and Reprogramming Cellular Energy Metabolism during the Fibrosis Process. Int J Mol Sci 2017; 18:ijms18122537. [PMID: 29186898 PMCID: PMC5751140 DOI: 10.3390/ijms18122537] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 11/10/2017] [Accepted: 11/21/2017] [Indexed: 01/06/2023] Open
Abstract
Fibrosis is characterized by fibroblast proliferation and fibroblast differentiation into myofibroblasts, which generate a relaxation-free contraction mechanism associated with excessive collagen synthesis in the extracellular matrix, which promotes irreversible tissue retraction evolving towards fibrosis. From a thermodynamic point of view, the mechanisms leading to fibrosis are irreversible processes that can occur through changing the entropy production rate. The thermodynamic behaviors of metabolic enzymes involved in fibrosis are modified by the dysregulation of both transforming growth factor β (TGF-β) signaling and the canonical WNT/β-catenin pathway, leading to aerobic glycolysis, called the Warburg effect. Molecular signaling pathways leading to fibrosis are considered dissipative structures that exchange energy or matter with their environment far from the thermodynamic equilibrium. The myofibroblastic cells arise from exergonic processes by switching the core metabolism from oxidative phosphorylation to glycolysis, which generates energy and reprograms cellular energy metabolism to induce the process of myofibroblast differentiation. Circadian rhythms are far-from-equilibrium thermodynamic processes. They directly participate in regulating the TGF-β and WNT/β-catenin pathways involved in energetic dysregulation and enabling fibrosis. The present review focusses on the thermodynamic implications of the reprogramming of cellular energy metabolism, leading to fibroblast differentiation into myofibroblasts through the positive interplay between TGF-β and WNT/β-catenin pathways underlying in fibrosis.
Collapse
Affiliation(s)
- Alexandre Vallée
- Laboratory of Mathematics and Applications (LMA), DACTIM, UMR CNRS 7348, CHU de Poitiers and University of Poitiers, 86021 Poitiers, France.
| | - Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l'Est Francilien (GHEF), 77100 Meaux, France.
| | - Jean-Noël Vallée
- Laboratory of Mathematics and Applications (LMA), DACTIM, UMR CNRS 7348, CHU de Poitiers and University of Poitiers, 86021 Poitiers, France.
- CHU Amiens Picardie, University of Picardie Jules Verne (UPJV), 80025 Amiens, France.
| |
Collapse
|
199
|
Chen YJ, Huang BY, Yang CN. Molecular modeling on HIF-2α-ARNT dimer destabilization caused by R171A and/or V192D mutations in HIF-2α. J Mol Graph Model 2017; 79:35-45. [PMID: 29132019 DOI: 10.1016/j.jmgm.2017.10.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 10/12/2017] [Accepted: 10/12/2017] [Indexed: 10/18/2022]
Abstract
Oxygen homeostasis in normal and tumor cells is mediated by hypoxia-inducible factors (HIFs), which are active as heterodimer complexes, such as HIF-2α-aryl hydrocarbon receptor nuclear translocator (ARNT) and HIF-1α-ARNT. A series of mutations on the interfaces between HIF-2α and ARNT and on the domain-domain interface within HIF-2α has been reported to exert varying effects on HIF-2α-ARNT dimerization. In the present study, molecular dynamic simulations were conducted to evaluate HIF-2α mutations, namely R171A, V192D, and R171A/V192D, which are not involved in the interaction with ARNT but impede HIF-2α-ARNT dimerization. Our results indicate that these mutations induct local conformation leading to a shortened (by V192D) or widened (by R171A and R171A/V192D) Y91-E346 separation distance, where E346 and Y91 are located on the HIF-2α and interact with ARNT according to electrostatic and geometrical shape complementarity, respectively.
Collapse
Affiliation(s)
- Ya-Jyun Chen
- Department of Life Sciences, National University of Kaohsiung, Kaohsiung, Taiwan
| | - Bo-Yen Huang
- Department of Life Sciences, National University of Kaohsiung, Kaohsiung, Taiwan
| | - Chia-Ning Yang
- Department of Life Sciences, National University of Kaohsiung, Kaohsiung, Taiwan; Scientific Multi-Simulation Center, National University of Kaohsiung, Kaohsiung, Taiwan.
| |
Collapse
|
200
|
Singh D, Arora R, Kaur P, Singh B, Mannan R, Arora S. Overexpression of hypoxia-inducible factor and metabolic pathways: possible targets of cancer. Cell Biosci 2017; 7:62. [PMID: 29158891 PMCID: PMC5683220 DOI: 10.1186/s13578-017-0190-2] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 11/06/2017] [Indexed: 12/18/2022] Open
Abstract
Cancer, the main cause of human deaths in the modern world is a group of diseases. Anticancer drug discovery is a challenge for scientists because of involvement of multiple survival pathways of cancer cells. An extensive study on the regulation of each step of these pathways may help find a potential cancer target. Up-regulated HIF-1 expression and altered metabolic pathways are two classical characteristics of cancer. Oxygen-dependent (through pVHL, PHDs, calcium-mediated) and independent (through growth factor signaling pathway, mdm2 pathway, HSP90) regulation of HIF-1α leads to angiogenesis, metastasis, and cell survival. The two subunits of HIF-1 regulates in the same fashion through different mechanisms. HIF-1α translation upregulates via mammalian target of rapamycin and mitogen-activated protein kinase signaling pathways, whereas HIF-1β through calmodulin kinase. Further, the stabilized interactions of these two subunits are important for proper functioning. Also, metabolic pathways crucial for the formation of building blocks (pentose phosphate pathway) and energy generation (glycolysis, TCA cycle and catabolism of glutamine) are altered in cancer cells to protect them from oxidative stress and to meet the reduced oxygen and nutrient supply. Up-regulated anaerobic metabolism occurs through enhanced expression of hexokinase, phosphofructokinase, triosephosphate isomerase, glucose 6-phosphate dehydrogenase and down-regulation of aerobic metabolism via pyruvate dehydrogenase kinase and lactate dehydrogenase which compensate energy requirements along with high glucose intake. Controlled expression of these two pathways through their common intermediate may serve as potent cancer target in future.
Collapse
Affiliation(s)
- Davinder Singh
- Department of Botanical and Environmental Sciences, Guru Nanak Dev University, Amritsar, 143005 India
| | - Rohit Arora
- Department of Biochemistry, Sri Guru Ram Das University of Health Sciences, Amritsar, 143001 India
| | - Pardeep Kaur
- Department of Botanical and Environmental Sciences, Guru Nanak Dev University, Amritsar, 143005 India
| | - Balbir Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005 India
| | - Rahul Mannan
- Department of Pathology, Sri Guru Ram Das University of Health Sciences, Amritsar, 143001 India
| | - Saroj Arora
- Department of Botanical and Environmental Sciences, Guru Nanak Dev University, Amritsar, 143005 India
| |
Collapse
|