1
|
Koltai T, Fliegel L. Dichloroacetate for Cancer Treatment: Some Facts and Many Doubts. Pharmaceuticals (Basel) 2024; 17:744. [PMID: 38931411 PMCID: PMC11206832 DOI: 10.3390/ph17060744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/23/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Rarely has a chemical elicited as much controversy as dichloroacetate (DCA). DCA was initially considered a dangerous toxic industrial waste product, then a potential treatment for lactic acidosis. However, the main controversies started in 2008 when DCA was found to have anti-cancer effects on experimental animals. These publications showed contradictory results in vivo and in vitro such that a thorough consideration of this compound's in cancer is merited. Despite 50 years of experimentation, DCA's future in therapeutics is uncertain. Without adequate clinical trials and health authorities' approval, DCA has been introduced in off-label cancer treatments in alternative medicine clinics in Canada, Germany, and other European countries. The lack of well-planned clinical trials and its use by people without medical training has discouraged consideration by the scientific community. There are few thorough clinical studies of DCA, and many publications are individual case reports. Case reports of DCA's benefits against cancer have been increasing recently. Furthermore, it has been shown that DCA synergizes with conventional treatments and other repurposable drugs. Beyond the classic DCA target, pyruvate dehydrogenase kinase, new target molecules have also been recently discovered. These findings have renewed interest in DCA. This paper explores whether existing evidence justifies further research on DCA for cancer treatment and it explores the role DCA may play in it.
Collapse
Affiliation(s)
- Tomas Koltai
- Hospital del Centro Gallego de Buenos Aires, Buenos Aires 2199, Argentina
| | - Larry Fliegel
- Department of Biochemistry, University Alberta, Edmonton, AB T6G 2H7, Canada;
| |
Collapse
|
2
|
Stacpoole PW, McCall CE. The pyruvate dehydrogenase complex: Life's essential, vulnerable and druggable energy homeostat. Mitochondrion 2023; 70:59-102. [PMID: 36863425 DOI: 10.1016/j.mito.2023.02.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 01/30/2023] [Accepted: 02/13/2023] [Indexed: 03/04/2023]
Abstract
Found in all organisms, pyruvate dehydrogenase complexes (PDC) are the keystones of prokaryotic and eukaryotic energy metabolism. In eukaryotic organisms these multi-component megacomplexes provide a crucial mechanistic link between cytoplasmic glycolysis and the mitochondrial tricarboxylic acid (TCA) cycle. As a consequence, PDCs also influence the metabolism of branched chain amino acids, lipids and, ultimately, oxidative phosphorylation (OXPHOS). PDC activity is an essential determinant of the metabolic and bioenergetic flexibility of metazoan organisms in adapting to changes in development, nutrient availability and various stresses that challenge maintenance of homeostasis. This canonical role of the PDC has been extensively probed over the past decades by multidisciplinary investigations into its causal association with diverse physiological and pathological conditions, the latter making the PDC an increasingly viable therapeutic target. Here we review the biology of the remarkable PDC and its emerging importance in the pathobiology and treatment of diverse congenital and acquired disorders of metabolic integration.
Collapse
Affiliation(s)
- Peter W Stacpoole
- Department of Medicine (Division of Endocrinology, Metabolism and Diabetes), and Department of Biochemistry and Molecular Biology, University of Florida, College of Medicine, Gainesville, FL, United States.
| | - Charles E McCall
- Department of Internal Medicine and Translational Sciences, and Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
3
|
Targeting hypoxia-related metabolism molecules: How to improve tumour immune and clinical treatment? Biomed Pharmacother 2022; 156:113917. [DOI: 10.1016/j.biopha.2022.113917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/21/2022] [Accepted: 10/24/2022] [Indexed: 11/20/2022] Open
|
4
|
Tiersma JF, Evers B, Bakker BM, Jalving M, de Jong S. Pyruvate Dehydrogenase Kinase Inhibition by Dichloroacetate in Melanoma Cells Unveils Metabolic Vulnerabilities. Int J Mol Sci 2022; 23:ijms23073745. [PMID: 35409102 PMCID: PMC8999016 DOI: 10.3390/ijms23073745] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/21/2022] [Accepted: 03/25/2022] [Indexed: 12/03/2022] Open
Abstract
Melanoma is characterized by high glucose uptake, partially mediated through elevated pyruvate dehydrogenase kinase (PDK), making PDK a potential treatment target in melanoma. We aimed to reduce glucose uptake in melanoma cell lines through PDK inhibitors dichloroacetate (DCA) and AZD7545 and through PDK knockdown, to inhibit cell growth and potentially unveil metabolic co-vulnerabilities resulting from PDK inhibition. MeWo cells were most sensitive to DCA, while SK-MEL-2 was the least sensitive, with IC50 values ranging from 13.3 to 27.0 mM. DCA strongly reduced PDH phosphorylation and increased the oxygen consumption rate:extracellular acidification rate (OCR:ECAR) ratio up to 6-fold. Knockdown of single PDK isoforms had similar effects on PDH phosphorylation and OCR:ECAR ratio as DCA but did not influence sensitivity to DCA. Growth inhibition by DCA was synergistic with the glutaminase inhibitor CB-839 (2- to 5-fold sensitization) and with diclofenac, known to inhibit monocarboxylate transporters (MCTs) (3- to 8-fold sensitization). CB-839 did not affect the OCR:ECAR response to DCA, whereas diclofenac strongly inhibited ECAR and further increased the OCR:ECAR ratio. We conclude that in melanoma cell lines, DCA reduces proliferation through reprogramming of cellular metabolism and synergizes with other metabolically targeted drugs.
Collapse
Affiliation(s)
- Jiske F. Tiersma
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands;
| | - Bernard Evers
- Laboratory of Pediatrics, Section Systems Medicine of Metabolism and Signalling, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (B.E.); (B.M.B.)
| | - Barbara M. Bakker
- Laboratory of Pediatrics, Section Systems Medicine of Metabolism and Signalling, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (B.E.); (B.M.B.)
| | - Mathilde Jalving
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands;
- Correspondence: (M.J.); (S.d.J.); Tel.: +31-50-3615692 (M.J.); +31-50-3612964 (S.d.J.)
| | - Steven de Jong
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands;
- Correspondence: (M.J.); (S.d.J.); Tel.: +31-50-3615692 (M.J.); +31-50-3612964 (S.d.J.)
| |
Collapse
|
5
|
Zhou Y, Guo Y, Tam KY. Targeting glucose metabolism to develop anticancer treatments and therapeutic patents. Expert Opin Ther Pat 2022; 32:441-453. [PMID: 35001793 DOI: 10.1080/13543776.2022.2027912] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION One of the most distinctive hallmarks of cancer cells is increased glucose consumption for aerobic glycolysis which is named the Warburg effect. In recent decades, extensive research has been carried out to exploit this famous phenomenon, trying to detect promising targetable vulnerabilities in altered metabolism to fight cancer. Targeting aberrant glucose metabolism can perturb cancer malignant proliferation and even induce programmed cell death. AREAS COVERED This review covered the recent patents which focused on targeting key glycolytic enzymes including hexokinase, pyruvate dehydrogenase kinases and lactate dehydrogenase for cancer treatment. EXPERT OPINION Compared with the conventional cancer treatment, specifically targeting the well-known Achilles heel Warburg effect has attracted considerable attention. Although there is still no single glycolytic agent for clinical cancer treatment, the combination of glycolytic inhibitor with conventional anticancer drug or the combined use of multiple glycolytic inhibitors are being investigated extensively in recent years, which could emerge as attractive anticancer strategies.
Collapse
Affiliation(s)
- Yan Zhou
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR, PR China
| | - Yizhen Guo
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR, PR China
| | - Kin Yip Tam
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR, PR China
| |
Collapse
|
6
|
Metformin and sodium dichloroacetate effects on proliferation, apoptosis, and metabolic activity tested alone and in combination in a canine prostate and a bladder cancer cell line. PLoS One 2021; 16:e0257403. [PMID: 34570803 PMCID: PMC8476037 DOI: 10.1371/journal.pone.0257403] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 08/31/2021] [Indexed: 01/26/2023] Open
Abstract
An important approach in tumor therapy is combining substances with different action mechanisms aiming to enhance the antineoplastic effect, decrease the therapeutic dosage, and avoid resistance mechanisms. Moreover, evaluating compounds already approved for the treatment of non-neoplastic diseases is promising for new antineoplastic therapies. Sodium dichloroacetate (DCA) reactivates oxidative phosphorylation in the cancer cell mitochondria, reducing apoptosis resistance in cancer cells. Furthermore, metformin inhibits the proliferation of tumor cells and CD133+ cancer -stem-like cells. In the present study, we evaluated the independent and synergistic effect of metformin and DCA on the metabolic activity, cell proliferation, and apoptosis of a canine prostate adenocarcinoma (Adcarc1258) and a transitional cell carcinoma cell line (TCC1506) in comparison to a primary canine fibroblast culture. Determining metformin uptake in tumor cells was performed by quantitative HPLC. Depending on the dosage, metformin as a single agent inhibited the metabolic activity and cell proliferation of the tumor cells, showing only minor effects on the fibroblasts. Furthermore, 1 mM metformin increased apoptosis over 96 h in the tumor cell lines but not in fibroblasts. Additionally, metformin uptake into the tumor cells in vitro was measurable by quantitative HPLC. Synergistic effects for the combination therapy were observed in both neoplastic cell lines as well as in the fibroblasts. Based on these results, metformin might be a promising therapeutic agent for canine urogenital tumors. Further studies on kinetics, toxicology, bioavailability, and application of metformin in dogs are necessary.
Collapse
|
7
|
Kim TS, Lee M, Park M, Kim SY, Shim MS, Lee CY, Choi DH, Cho Y. Metformin and Dichloroacetate Suppress Proliferation of Liver Cancer Cells by Inhibiting mTOR Complex 1. Int J Mol Sci 2021; 22:ijms221810027. [PMID: 34576192 PMCID: PMC8467948 DOI: 10.3390/ijms221810027] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 12/15/2022] Open
Abstract
The Warburg effect is important for cancer cell proliferation. This phenomenon can be flexible by interaction between glycolysis and mitochondrial oxidation for energy production. We aimed to investigate the anticancer effects of the pyruvate dehydrogenase kinase inhibitor, dichloroacetate (DCA) and the mitochondrial respiratory complex I inhibitor metformin in liver cancer cells. The anticancer effect of DCA and/or metformin on HepG2, PLC/PRF5 human liver cancer cell lines, MH-134 murine hepatoma cell lines, and primary normal hepatocytes using MTT assay. Inhibition of lactate/ATP production and intracellular reactive oxygen species generation by DCA and metformin was investigated. Inhibition of PI3K/Akt/mTOR complex I was evaluated to see whether it occurred through AMPK signaling. Anticancer effects of a combination treatment of DCA and metformin were evaluated in HCC murine model. The results showed that metformin and DCA effectively induced apoptosis in liver cancer cells. A combination treatment of metformin and DCA did not affect viability of primary normal hepatocytes. Metformin upregulated glycolysis in liver cancer cells, thereby increasing sensitivity to the DCA treatment. Metformin and DCA inhibited mTOR complex I signaling through upregulated AMPK-independent REDD1. In addition, metformin and DCA increased reactive oxygen species levels in liver cancer cells, which induced apoptosis. A combination treatment of metformin and DCA significantly suppressed the tumor growth of liver cancer cells using in vivo xenograft model. Taken together, the combined treatment of metformin and DCA suppressed the growth of liver cancer cells. This strategy may be effective for patients with advanced liver cancer.
Collapse
Affiliation(s)
- Tae Suk Kim
- Department of Internal Medicine, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon 24341, Korea; (T.S.K.); (C.Y.L.); (D.H.C.)
| | - Minjong Lee
- Department of Internal Medicine, Ewha Womans University College of Medicine, Seoul 07804, Korea;
- Department of Internal Medicine, Ewha Womans University Medical Center, Seoul 07804, Korea
- Correspondence: (M.L.); (Y.C.); Tel.: +82-2-6986-1761 (M.L.); +82-31-920-1605 (Y.C.)
| | - Minji Park
- Department of Internal Medicine, Ewha Womans University College of Medicine, Seoul 07804, Korea;
- Department of Internal Medicine, Ewha Womans University Medical Center, Seoul 07804, Korea
- Department of Internal Medicine, CHA Gangnam Medical Center, CHA University School of Medicine, Seoul 06135, Korea
| | - Sae Yun Kim
- Department of Pediatrics, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| | - Min Suk Shim
- Division of Bioengineering, Incheon National University, Incheon 22012, Korea;
| | - Chea Yeon Lee
- Department of Internal Medicine, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon 24341, Korea; (T.S.K.); (C.Y.L.); (D.H.C.)
| | - Dae Hee Choi
- Department of Internal Medicine, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon 24341, Korea; (T.S.K.); (C.Y.L.); (D.H.C.)
| | - Yuri Cho
- Department of Internal Medicine, CHA Gangnam Medical Center, CHA University School of Medicine, Seoul 06135, Korea
- Center for Liver and Pancreatobiliary Cancer, Research Institute and Hospital, National Cancer Center, Goyang 10408, Korea
- Correspondence: (M.L.); (Y.C.); Tel.: +82-2-6986-1761 (M.L.); +82-31-920-1605 (Y.C.)
| |
Collapse
|
8
|
Suppression of Pyruvate Dehydrogenase Kinase by Dichloroacetate in Cancer and Skeletal Muscle Cells Is Isoform Specific and Partially Independent of HIF-1α. Int J Mol Sci 2021; 22:ijms22168610. [PMID: 34445316 PMCID: PMC8395311 DOI: 10.3390/ijms22168610] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/28/2021] [Accepted: 08/03/2021] [Indexed: 02/01/2023] Open
Abstract
Inhibition of pyruvate dehydrogenase kinase (PDK) emerged as a potential strategy for treatment of cancer and metabolic disorders. Dichloroacetate (DCA), a prototypical PDK inhibitor, reduces the abundance of some PDK isoenzymes. However, the underlying mechanisms are not fully characterized and may differ across cell types. We determined that DCA reduced the abundance of PDK1 in breast (MDA-MB-231) and prostate (PC-3) cancer cells, while it suppressed both PDK1 and PDK2 in skeletal muscle cells (L6 myotubes). The DCA-induced PDK1 suppression was partially dependent on hypoxia-inducible factor-1α (HIF-1α), a transcriptional regulator of PDK1, in cancer cells but not in L6 myotubes. However, the DCA-induced alterations in the mRNA and the protein levels of PDK1 and/or PDK2 did not always occur in parallel, implicating a role for post-transcriptional mechanisms. DCA did not inhibit the mTOR signaling, while inhibitors of the proteasome or gene silencing of mitochondrial proteases CLPP and AFG3L2 did not prevent the DCA-induced reduction of the PDK1 protein levels. Collectively, our results suggest that DCA reduces the abundance of PDK in an isoform-dependent manner via transcriptional and post-transcriptional mechanisms. Differential response of PDK isoenzymes to DCA might be important for its pharmacological effects in different types of cells.
Collapse
|
9
|
Korsakova L, Krasko JA, Stankevicius E. Metabolic-targeted Combination Therapy With Dichloroacetate and Metformin Suppresses Glioblastoma Cell Line Growth In Vitro and In Vivo. In Vivo 2021; 35:341-348. [PMID: 33402483 DOI: 10.21873/invivo.12265] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND/AIM We investigated the hypothesis that dichloroacetate (DCA), a pyruvate dehydrogenase kinase inhibitor, and metformin (MET), an antidiabetic agent and complex I inhibitor, have synergistic cytotoxic effects in glioblastoma cells in vitro and in vivo. MATERIALS AND METHODS We performed dose response experiments and combination index calculation. Apoptotic and necrotic cells were estimated by flow cytometry. Cell metabolism was evaluated by Seahorse analysis and lactate export. Overall survival and tumor volume growth experiments were performed in C57BL/6 mice GL-261 allograft model. RESULTS DCA and MET showed dose-dependent cytotoxicity and synergistic effects. DCA alleviated the increase in lactate production induced by MET. Seahorse analysis showed that DCA treatment results in increased oxygen consumption rate, which is decreased by MET. DCA and MET significantly inhibited tumor growth and increased overall survival in mice. CONCLUSION Compounds targeting tumor cell metabolism could become potential treatment options for glioblastoma multiforme.
Collapse
Affiliation(s)
- Laura Korsakova
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania;
| | | | - Edgaras Stankevicius
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania;
| |
Collapse
|
10
|
Sobiepanek A, Paone A, Cutruzzolà F, Kobiela T. Biophysical characterization of melanoma cell phenotype markers during metastatic progression. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2021; 50:523-542. [PMID: 33730175 PMCID: PMC8190004 DOI: 10.1007/s00249-021-01514-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/30/2021] [Accepted: 03/08/2021] [Indexed: 12/14/2022]
Abstract
Melanoma is the most fatal form of skin cancer, with increasing prevalence worldwide. The most common melanoma genetic driver is mutation of the proto-oncogene serine/threonine kinase BRAF; thus, the inhibition of its MAP kinase pathway by specific inhibitors is a commonly applied therapy. However, many patients are resistant, or develop resistance to this type of monotherapy, and therefore combined therapies which target other signaling pathways through various molecular mechanisms are required. A possible strategy may involve targeting cellular energy metabolism, which has been recognized as crucial for cancer development and progression and which connects through glycolysis to cell surface glycan biosynthetic pathways. Protein glycosylation is a hallmark of more than 50% of the human proteome and it has been recognized that altered glycosylation occurs during the metastatic progression of melanoma cells which, in turn facilitates their migration. This review provides a description of recent advances in the search for factors able to remodel cell metabolism between glycolysis and oxidative phosphorylation, and of changes in specific markers and in the biophysical properties of cells during melanoma development from a nevus to metastasis. This development is accompanied by changes in the expression of surface glycans, with corresponding changes in ligand-receptor affinity, giving rise to structural features and viscoelastic parameters particularly well suited to study by label-free biophysical methods.
Collapse
Affiliation(s)
- Anna Sobiepanek
- Laboratory of Biomolecular Interactions Studies, Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664, Warsaw, Poland.
| | - Alessio Paone
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Francesca Cutruzzolà
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Tomasz Kobiela
- Laboratory of Biomolecular Interactions Studies, Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664, Warsaw, Poland
| |
Collapse
|
11
|
Paredes F, Williams HC, San Martin A. Metabolic adaptation in hypoxia and cancer. Cancer Lett 2021; 502:133-142. [PMID: 33444690 PMCID: PMC8158653 DOI: 10.1016/j.canlet.2020.12.020] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 12/09/2020] [Accepted: 12/11/2020] [Indexed: 12/17/2022]
Abstract
The ability of tumor cells to adapt to changes in oxygen tension is essential for tumor development. Low oxygen concentration influences cellular metabolism and, thus, affects proliferation, migration, and invasion. A focal point of the cell's adaptation to hypoxia is the transcription factor HIF1α (hypoxia-inducible factor 1 alpha), which affects the expression of specific gene networks involved in cellular energetics and metabolism. This review illustrates the mechanisms by which HIF1α-induced metabolic adaptation promotes angiogenesis, participates in the escape from immune recognition, and increases cancer cell antioxidant capacity. In addition to hypoxia, metabolic inhibition of 2-oxoglutarate-dependent dioxygenases regulates HIF1α stability and transcriptional activity. This phenomenon, known as pseudohypoxia, is frequently used by cancer cells to promote glycolytic metabolism to support biomass synthesis for cell growth and proliferation. In this review, we highlight the role of the most important metabolic intermediaries that are at the center of cancer's biology, and in particular, the participation of these metabolites in HIF1α retrograde signaling during the establishment of pseudohypoxia. Finally, we will discuss how these changes affect both the development of cancers and their resistance to treatment.
Collapse
Affiliation(s)
- Felipe Paredes
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, GA, 30322, USA
| | - Holly C Williams
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, GA, 30322, USA
| | - Alejandra San Martin
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
12
|
Blocking Aerobic Glycolysis by Targeting Pyruvate Dehydrogenase Kinase in Combination with EGFR TKI and Ionizing Radiation Increases Therapeutic Effect in Non-Small Cell Lung Cancer Cells. Cancers (Basel) 2021; 13:cancers13050941. [PMID: 33668151 PMCID: PMC7956357 DOI: 10.3390/cancers13050941] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/16/2021] [Accepted: 02/19/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Non-small cell lung cancer (NSCLC) patients harboring oncogenic mutations in the epidermal growth factor receptor (EGFR) inevitably develop resistance to targeted EGFR tyrosine kinase inhibitors (TKI) therapy. To support malignant features associated with cancer development and therapy resistance, the cancer cells adapt their metabolic rate and pathways. As an example, aerobic glycolysis, where the cells use glycolysis in the presence of oxygen, is frequently seen. Here we show that targeting aerobic glycolysis represents a promising strategy in cancer therapeutics. Abstract Increased glycolytic activity is a hallmark of cancer initiation and progression and is often observed in non-small cell lung cancer (NSCLC). Pyruvate dehydrogenase (PDH) complex acts as a gatekeeper between glycolysis and oxidative phosphorylation, and activation of PDH is known to inhibit glycolytic activity. As part of a standard therapeutic regimen, patients with NSCLC harboring oncogenic mutations in the epidermal growth factor receptor (EGFR) are treated with EGFR tyrosine kinase inhibitors (EGFR TKIs). Independent of good initial response, development of resistance to this therapy is inevitable. In the presented work, we propose that inhibition of glycolysis will add to the therapeutic effects and possibly prevent development of resistance against both EGFR TKIs and ionizing radiation in NSCLC. Analysis of transcriptome data from two independent NSCLC patient cohorts identified increased expression of pyruvate dehydrogenase kinase 1 (PDHK1) as well as upregulated expression of genes involved in glucose metabolism in tumors compared to normal tissue. We established in vitro models of development of resistance to EGFR TKIs to study metabolism and determine if targeting PDHK would prevent development of resistance to EGFR TKIs in NSCLC cells. The PDHK1 inhibitor dichloroacetate (DCA) in combination with EGFR TKIs and/or ionizing radiation was shown to increase the therapeutic effect in our NSCLC cell models. This mechanism was associated with redirected metabolism towards pyruvate oxidation and reduced lactate production, both in EGFR TKI sensitive and resistant NSCLC cells. Using DCA, the intracellular pool of pyruvate available for lactic fermentation becomes limited. Consequently, pyruvate is redirected to the mitochondria, and reinforces mitochondrial activity. Addition of DCA to cell culture deacidifies the extracellular microenvironment as less lactate is produced and excreted. In our study, we find that this redirection of metabolism adds to the therapeutic effect of EGFR TKI and ionizing radiation in NSCLC.
Collapse
|
13
|
Jiang Y, Chen P, Hu K, Dai G, Li J, Zheng D, Yuan H, He L, Xie P, Tu M, Peng S, Qu C, Lin W, Chung RT, Hong J. Inflammatory microenvironment of fibrotic liver promotes hepatocellular carcinoma growth, metastasis and sorafenib resistance through STAT3 activation. J Cell Mol Med 2021; 25:1568-1582. [PMID: 33410581 PMCID: PMC7875922 DOI: 10.1111/jcmm.16256] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 10/14/2019] [Accepted: 12/14/2020] [Indexed: 12/12/2022] Open
Abstract
The pro‐inflammatory and pro‐fibrotic liver microenvironment facilitates hepatocarcinogenesis. However, the effects and mechanisms by which the hepatic fibroinflammatory microenvironment modulates intrahepatic hepatocellular carcinoma (HCC) progression and its response to systematic therapy remain largely unexplored. We established a syngeneic orthotopic HCC mouse model with a series of persistent liver injury induced by CCl4 gavage, which mimic the dynamic effect of hepatic pathology microenvironment on intrahepatic HCC growth and metastasis. Non‐invasive bioluminescence imaging was applied to follow tumour progression over time. The effect of the liver microenvironment modulated by hepatic injury on sorafenib resistance was investigated in vivo and in vitro. We found that the persistent liver injury facilitated HCC growth and metastasis, which was positively correlated with the degree of liver inflammation rather than the extent of liver fibrosis. The inflammatory cytokines in liver tissue were clearly increased after liver injury. The two indicated cytokines, tumour necrosis factor‐α (TNF‐α) and interleukin‐6 (IL‐6), both promoted intrahepatic HCC progression via STAT3 activation. In addition, the hepatic inflammatory microenvironment contributed to sorafenib resistance through the anti‐apoptotic protein mediated by STAT3, and STAT3 inhibitor S3I‐201 significantly improved sorafenib efficacy impaired by liver inflammation. Clinically, the increased inflammation of liver tissues was accompanied with the up‐regulated STAT3 activation in HCC. Above all, we concluded that the hepatic inflammatory microenvironment promotes intrahepatic HCC growth, metastasis and sorafenib resistance through activation of STAT3.
Collapse
Affiliation(s)
- Yuchuan Jiang
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Peng Chen
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Kaishun Hu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Guanqi Dai
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Jinying Li
- Department of Gastroenterology, Guangzhou Overseas Chinese Hospital, Jinan University, Guangzhou, China
| | - Dandan Zheng
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Hui Yuan
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Lu He
- Department of Radiotherapy, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Penghui Xie
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Mengxian Tu
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Shuang Peng
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, China
| | - Chen Qu
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Wenyu Lin
- Liver Center and Gastrointestinal Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Raymond T Chung
- Liver Center and Gastrointestinal Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jian Hong
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
14
|
Cioce M, Pulito C, Strano S, Blandino G, Fazio VM. Metformin: Metabolic Rewiring Faces Tumor Heterogeneity. Cells 2020; 9:E2439. [PMID: 33182253 PMCID: PMC7695274 DOI: 10.3390/cells9112439] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 10/13/2020] [Accepted: 11/03/2020] [Indexed: 02/07/2023] Open
Abstract
Tumor heterogeneity impinges on all the aspects of tumor history, from onset to metastasis and relapse. It is growingly recognized as a propelling force for tumor adaptation to environmental and micro-environmental cues. Metabolic heterogeneity perfectly falls into this process. It strongly contributes to the metabolic plasticity which characterizes cancer cell subpopulations-capable of adaptive switching under stress conditions, between aerobic glycolysis and oxidative phosphorylation-in both a convergent and divergent modality. The mitochondria appear at center-stage in this adaptive process and thus, targeting mitochondria in cancer may prove of therapeutic value. Metformin is the oldest and most used anti-diabetic medication and its relationship with cancer has witnessed rises and falls in the last 30 years. We believe it is useful to revisit the main mechanisms of action of metformin in light of the emerging views on tumor heterogeneity. We first analyze the most consolidated view of its mitochondrial mechanism of action and then we frame the latter in the context of tumor adaptive strategies, cancer stem cell selection, metabolic zonation of tumors and the tumor microenvironment. This may provide a more critical point of view and, to some extent, may help to shed light on some of the controversial evidence for metformin's anticancer action.
Collapse
Affiliation(s)
- Mario Cioce
- Department of Medicine, R.U. in Molecular Medicine and Biotechnology, University Campus Bio-Medico of Rome, 00128 Rome, Italy;
| | - Claudio Pulito
- Oncogenomic and Epigenetic Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (C.P.); (G.B.)
| | - Sabrina Strano
- SAFU Unit, Department of Research, Diagnosis and Innovative Technologies, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Giovanni Blandino
- Oncogenomic and Epigenetic Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (C.P.); (G.B.)
| | - Vito Michele Fazio
- Department of Medicine, R.U. in Molecular Medicine and Biotechnology, University Campus Bio-Medico of Rome, 00128 Rome, Italy;
- Institute of Translation Pharmacology, National Research Council of Italy (CNR), 00133 Rome, Italy
| |
Collapse
|
15
|
Cui Y, Zhou J, Rong F. Combination of metformin and RG7388 enhances inhibition of growth and induction of apoptosis of ovarian cancer cells through the PI3K/AKT/mTOR pathway. Biochem Biophys Res Commun 2020; 533:665-671. [PMID: 33051060 DOI: 10.1016/j.bbrc.2020.09.135] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 09/29/2020] [Indexed: 10/23/2022]
Abstract
Ovarian cancer is a gynecological cancer that has the highest mortality rate and is often resistant to conventional treatments. Therefore, development of new therapies is essential. Metformin (MET), which is the priority drug for treatment of type 2 diabetes, has received increasing attention because of its anti-tumor effects. Here, we examined combined anti-tumor effects of MET and RG7388, the only MDM2 (mouse double minute 2 homolog) antagonist that has entered phase III clinical trials, on ovarian cancer cell lines. We examined effects on proliferation by Cell Counting Kit-8 (CCK-8) and colony formation assays, and effects on apoptosis by flow cytometric analysis and Hoechst staining. Western blotting was used to measure protein expression in cells and tissues treated with MET and/or RG7388. Flow cytometry was used to measure reactive oxygen species (ROS). We also examined the effects of MET and/or RG7388 on inhibition of A2780 cell growth in vivo. The combination of MET and RG7388 significantly increased growth inhibition, apoptosis, and ROS of A2780 and SKOV3 cells compared with either agent alone. Additionally, in vitro and in vivo results showed that MET and/or RG7388 inhibited the phosphoinositide 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathway and their combination had a stronger effect. Our findings suggest that the combination of MET and RG7388 enhances growth inhibition and apoptosis induction of ovarian cancer cells through the PI3K/AKT/mTOR pathway and accumulation of intracellular ROS.
Collapse
Affiliation(s)
- Yingying Cui
- Department of Obstetrics and Gynecology, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250014, China
| | - Jing Zhou
- Department of Obstetrics and Gynecology, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250014, China
| | - Fengnian Rong
- Department of Obstetrics and Gynecology, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250014, China.
| |
Collapse
|
16
|
Ruhnau J, Parczyk J, Danker K, Eickholt B, Klein A. Synergisms of genome and metabolism stabilizing antitumor therapy (GMSAT) in human breast and colon cancer cell lines: a novel approach to screen for synergism. BMC Cancer 2020; 20:617. [PMID: 32615946 PMCID: PMC7331156 DOI: 10.1186/s12885-020-07062-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 06/11/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Despite an improvement of prognosis in breast and colon cancer, the outcome of the metastatic disease is still severe. Microevolution of cancer cells often leads to drug resistance and tumor-recurrence. To target the driving forces of the tumor microevolution, we focused on synergistic drug combinations of selected compounds. The aim is to prevent the tumor from evolving in order to stabilize disease remission. To identify synergisms in a high number of compounds, we propose here a three-step concept that is cost efficient, independent of high-throughput machines and reliable in its predictions. METHODS We created dose response curves using MTT- and SRB-assays with 14 different compounds in MCF-7, HT-29 and MDA-MB-231 cells. In order to efficiently screen for synergies, we developed a screening tool in which 14 drugs were combined (91 combinations) in MCF-7 and HT-29 using EC25 or less. The most promising combinations were verified by the method of Chou and Talalay. RESULTS All 14 compounds exhibit antitumor effects on each of the three cell lines. The screening tool resulted in 19 potential synergisms detected in HT-29 (20.9%) and 27 in MCF-7 (29.7%). Seven of the top combinations were further verified over the whole dose response curve, and for five combinations a significant synergy could be confirmed. The combination Nutlin-3 (inhibition of MDM2) and PX-478 (inhibition of HIF-1α) could be confirmed for all three cell lines. The same accounts for the combination of Dichloroacetate (PDH activation) and NHI-2 (LDH-A inhibition). Our screening method proved to be an efficient tool that is reliable in its projections. CONCLUSIONS The presented three-step concept proved to be cost- and time-efficient with respect to the resulting data. The newly found combinations show promising results in MCF-7, HT-29 and MDA-MB231 cancer cells.
Collapse
Affiliation(s)
- Jérôme Ruhnau
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Biochemistry, Charitéplatz 1, 10117, Berlin, Germany.
| | - Jonas Parczyk
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Biochemistry, Charitéplatz 1, 10117, Berlin, Germany.
| | - Kerstin Danker
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Biochemistry, Charitéplatz 1, 10117, Berlin, Germany
| | - Britta Eickholt
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Biochemistry, Charitéplatz 1, 10117, Berlin, Germany
| | - Andreas Klein
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Biochemistry, Charitéplatz 1, 10117, Berlin, Germany
| |
Collapse
|
17
|
Skeberdytė A, Sarapinienė I, Krasko JA, Barakauskienė A, Žilionytė K, Prokarenkaitė R, Sužiedėlis K, Bukelskienė V, Jarmalaitė S. Salinomycin and dichloroacetate synergistically inhibit Lewis lung carcinoma cell proliferation, tumor growth and metastasis. Biochem Biophys Res Commun 2020; 523:874-879. [PMID: 31955885 DOI: 10.1016/j.bbrc.2019.12.107] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 12/21/2019] [Indexed: 12/27/2022]
Abstract
Drug combination is considered to be the cornerstone of cancer treatment. Simultaneous administration of two or more drugs but at lower doses not only increases cytotoxic effects on tumor cells, but also reduces side effects and possibly overcomes drug resistance. Salinomycin is a well-known cancer stem cell killer, and dichloroacetate is a pyruvate dehydrogenase kinase inhibitor that exclusively targets cells with altered mitochondrial activity, a characteristic being common to most of the cancer cells. In our recent study, we have demonstrated that salinomycin exerted a cytotoxic effect on colorectal carcinoma cells in the 2D and 3D cultures and provided evidence that the mechanism of their synergy was mediated by dichloroacetate-dependent inhibition of the activity of multidrug resistance proteins. In the current work, we confirmed the synergistic cytotoxic properties of salinomycin and dichloroacetate in the 2D and 3D cultures of Lewis lung carcinoma (LLC1) cells. To verify if a synergistic effect of these compounds persisted in vivo, we performed series of experiments using a syngeneic LLC1-C57BL/6 mouse model and demonstrated that combination therapy with salinomycin and DCA increased the survival rate of allografted mice, inhibited metastatic site formation and reduced the populations of cancer stem cells as well as cells that underwent the epithelial-to-mesenchymal transition. Our results demonstrate that a synergistic effect of salinomycin and dichloroacetate exists not only in vitro but also in vivo and suggest their benefits in the treatment of metastatic cancers.
Collapse
Affiliation(s)
- Aistė Skeberdytė
- Institute of Biosciences, Life Sciences Center, Vilnius University, Saulėtekio al. 7, LT-10222, Vilnius, Lithuania.
| | - Ieva Sarapinienė
- Institute of Cardiology, Lithuanian University of Health Sciences, Sukilėlių pr. 15, LT-50162, Kaunas, Lithuania
| | | | | | - Karolina Žilionytė
- National Cancer Institute, Santariškių g. 1, LT-08660, Vilnius, Lithuania
| | | | | | - Virginija Bukelskienė
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Saulėtekio al. 7, LT-10223, Vilnius, Lithuania
| | - Sonata Jarmalaitė
- National Cancer Institute, Santariškių g. 1, LT-08660, Vilnius, Lithuania
| |
Collapse
|
18
|
Mazurek M, Litak J, Kamieniak P, Kulesza B, Jonak K, Baj J, Grochowski C. Metformin as Potential Therapy for High-Grade Glioma. Cancers (Basel) 2020; 12:E210. [PMID: 31952173 PMCID: PMC7016983 DOI: 10.3390/cancers12010210] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 01/09/2020] [Accepted: 01/13/2020] [Indexed: 12/15/2022] Open
Abstract
Metformin (MET), 1,1-dimethylbiguanide hydrochloride, is a biguanide drug used as the first-line medication in the treatment of type 2 diabetes. The recent years have brought many observations showing metformin in its new role. The drug, commonly used in the therapy of diabetes, may also find application in the therapy of a vast variety of tumors. Its effectiveness has been demonstrated in colon, breast, prostate, pancreatic cancer, leukemia, melanoma, lung and endometrial carcinoma, as well as in gliomas. This is especially important in light of the poor options offered to patients in the case of high-grade gliomas, which include glioblastoma (GBM). A thorough understanding of the mechanism of action of metformin can make it possible to discover new drugs that could be used in neoplasm therapy.
Collapse
Affiliation(s)
- Marek Mazurek
- Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, Jaczewskiego 8, 20-954 Lublin, Poland; (M.M.); (J.L.); (P.K.); (B.K.)
| | - Jakub Litak
- Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, Jaczewskiego 8, 20-954 Lublin, Poland; (M.M.); (J.L.); (P.K.); (B.K.)
- Department of Immunology, Medical University of Lublin, Jaczewskiego 8, 20-954 Lublin, Poland
| | - Piotr Kamieniak
- Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, Jaczewskiego 8, 20-954 Lublin, Poland; (M.M.); (J.L.); (P.K.); (B.K.)
| | - Bartłomiej Kulesza
- Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, Jaczewskiego 8, 20-954 Lublin, Poland; (M.M.); (J.L.); (P.K.); (B.K.)
| | - Katarzyna Jonak
- Department of Foregin Languages, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland;
| | - Jacek Baj
- Department of Anatomy, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland;
| | - Cezary Grochowski
- Department of Anatomy, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland;
| |
Collapse
|
19
|
Schoonjans CA, Gallez B. Metabolic Plasticity of Tumor Cells: How They Do Adapt to Food Deprivation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1219:109-123. [PMID: 32130696 DOI: 10.1007/978-3-030-34025-4_6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Dysregulated metabolism is a key hallmark of cancer cells and an enticing target for cancer treatment. Since the last 10 years, research on cancer metabolism has moved from pathway attention to network consideration. This metabolic complexity continuously adapt to new constraints in the tumor microenvironment. In this review, we will highlight striking changes in cancer cell metabolism compared to normal cells. Understanding this tumor metabolic plasticity suggests potential new targets and innovative combinatorial treatments for fighting cancer.
Collapse
Affiliation(s)
- Céline A Schoonjans
- Université catholique de Louvain (UCLouvain), Louvain Drug Research Institute, Brussels, Belgium
| | | |
Collapse
|
20
|
Chu X, Schwartz R, Diamond MP, Raju RP. A Combination Treatment Strategy for Hemorrhagic Shock in a Rat Model Modulates Autophagy. Front Med (Lausanne) 2019; 6:281. [PMID: 31921865 PMCID: PMC6928057 DOI: 10.3389/fmed.2019.00281] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 11/18/2019] [Indexed: 12/11/2022] Open
Abstract
Hemorrhagic shock leads to whole body hypoxia and nutrient deprivation resulting in organ dysfunction and mortality. Previous studies demonstrated that resveratrol, dichloroacetate, and niacin improve organ function and survival in rats following hemorrhagic shock injury (HI). We hypothesized that a combinatorial formula that collectively promotes survival will decrease the dose of individual compounds toward effective therapy for HI. Sprague-Dawley rats were subjected to HI by withdrawing 60% blood volume. NiDaR (Niacin-Dichloroacetate-Resveratrol; 2 mg/kg dose of each) or vehicle was administered following the shock in the absence of fluid resuscitation, and survival monitored. In order to study alterations in molecular mediators, separate groups of rats were administered NiDaR or vehicle along with resuscitation fluid, following HI. We observed significant improvement (p < 0.05) in survival following HI in animals that received NiDaR, in the absence of fluid resuscitation. In NiDaR treated animals that received resuscitation fluid, MAP was significantly increased compared to Veh-treated rats. HI-induced increase in systemic IL-6 levels and tissue expression of IL-6, IL-10, IL-1β, and IL-18 genes in the heart were attenuated with NiDaR treatment. NiDaR promoted autophagy following HI as demonstrated by reduced p-mTOR, increased p-ULK1 and p-Beclin. The combinatorial formula, NiDaR, reduced inflammation, promoted autophagy, and reduced doses of individual compounds used, and may be more effective in genetically heterogeneous population. In conclusion our experiments demonstrated that the combinatorial drug treatment has salutary effect in rats following HI.
Collapse
Affiliation(s)
- Xiaogang Chu
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Richard Schwartz
- Emergency Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Michael P Diamond
- Department of Obstetrics and Gynaecology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Raghavan Pillai Raju
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
21
|
Stakišaitis D, Juknevičienė M, Damanskienė E, Valančiūtė A, Balnytė I, Alonso MM. The Importance of Gender-Related Anticancer Research on Mitochondrial Regulator Sodium Dichloroacetate in Preclinical Studies In Vivo. Cancers (Basel) 2019; 11:cancers11081210. [PMID: 31434295 PMCID: PMC6721567 DOI: 10.3390/cancers11081210] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 08/16/2019] [Accepted: 08/16/2019] [Indexed: 12/28/2022] Open
Abstract
Sodium dichloroacetate (DCA) is an investigational medicinal product which has a potential anticancer preparation as a metabolic regulator in cancer cells’ mitochondria. Inhibition of pyruvate dehydrogenase kinases by DCA keeps the pyruvate dehydrogenase complex in the active form, resulting in decreased lactic acid in the tumor microenvironment. This literature review displays the preclinical research data on DCA’s effects on the cell pyruvate dehydrogenase deficiency, pyruvate mitochondrial oxidative phosphorylation, reactive oxygen species generation, and the Na+–K+–2Cl− cotransporter expression regulation in relation to gender. It presents DCA pharmacokinetics and the hepatocarcinogenic effect, and the safety data covers the DCA monotherapy efficacy for various human cancer xenografts in vivo in male and female animals. Preclinical cancer researchers report the synergistic effects of DCA combined with different drugs on cancer by reversing resistance to chemotherapy and promoting cell apoptosis. Researchers note that female and male animals differ in the mechanisms of cancerogenesis but often ignore studying DCA’s effects in relation to gender. Preclinical gender-related differences in DCA pharmacology, pharmacological mechanisms, and the elucidation of treatment efficacy in gonad hormone dependency could be relevant for individualized therapy approaches so that gender-related differences in treatment response and safety can be proposed.
Collapse
Affiliation(s)
- Donatas Stakišaitis
- Laboratory of Molecular Oncology, National Cancer Institute, 08660 Vilnius, Lithuania.
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania.
| | - Milda Juknevičienė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Eligija Damanskienė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Angelija Valančiūtė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Ingrida Balnytė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Marta Maria Alonso
- Department of Pediatrics, Clínica Universidad de Navarra, University of Navarra, 55 Pamplona, Spain.
| |
Collapse
|
22
|
Aloia A, Müllhaupt D, Chabbert CD, Eberhart T, Flückiger-Mangual S, Vukolic A, Eichhoff O, Irmisch A, Alexander LT, Scibona E, Frederick DT, Miao B, Tian T, Cheng C, Kwong LN, Wei Z, Sullivan RJ, Boland GM, Herlyn M, Flaherty KT, Zamboni N, Dummer R, Zhang G, Levesque MP, Krek W, Kovacs WJ. A Fatty Acid Oxidation-dependent Metabolic Shift Regulates the Adaptation of BRAF-mutated Melanoma to MAPK Inhibitors. Clin Cancer Res 2019; 25:6852-6867. [PMID: 31375515 DOI: 10.1158/1078-0432.ccr-19-0253] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 05/23/2019] [Accepted: 07/30/2019] [Indexed: 12/11/2022]
Abstract
PURPOSE Treatment of BRAFV600E -mutant melanomas with MAPK inhibitors (MAPKi) results in significant tumor regression, but acquired resistance is pervasive. To understand nonmutational mechanisms underlying the adaptation to MAPKi and to identify novel vulnerabilities of melanomas treated with MAPKi, we focused on the initial response phase during treatment with MAPKi. EXPERIMENTAL DESIGN By screening proteins expressed on the cell surface of melanoma cells, we identified the fatty acid transporter CD36 as the most consistently upregulated protein upon short-term treatment with MAPKi. We further investigated the effects of MAPKi on fatty acid metabolism using in vitro and in vivo models and analyzing patients' pre- and on-treatment tumor specimens. RESULTS Melanoma cells treated with MAPKi displayed increased levels of CD36 and of PPARα-mediated and carnitine palmitoyltransferase 1A (CPT1A)-dependent fatty acid oxidation (FAO). While CD36 is a useful marker of melanoma cells during adaptation and drug-tolerant phases, the upregulation of CD36 is not functionally involved in FAO changes that characterize MAPKi-treated cells. Increased FAO is required for BRAFV600E -mutant melanoma cells to survive under the MAPKi-induced metabolic stress prior to acquiring drug resistance. The upfront and concomitant inhibition of FAO, glycolysis, and MAPK synergistically inhibits tumor cell growth in vitro and in vivo. CONCLUSIONS Thus, we identified a clinically relevant therapeutic approach that has the potential to improve initial responses and to delay acquired drug resistance of BRAFV600E -mutant melanoma.
Collapse
Affiliation(s)
- Andrea Aloia
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland.
| | - Daniela Müllhaupt
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | | | - Tanja Eberhart
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | | | - Ana Vukolic
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Ossia Eichhoff
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Anja Irmisch
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Leila T Alexander
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Ernesto Scibona
- Institute of Chemical and Bioengineering, ETH Zurich, Zurich, Switzerland
| | | | - Benchun Miao
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | - Tian Tian
- Department of Computer Science, New Jersey Institute of Technology, Newark, New Jersey
| | - Chaoran Cheng
- Department of Computer Science, New Jersey Institute of Technology, Newark, New Jersey
| | - Lawrence N Kwong
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zhi Wei
- Department of Computer Science, New Jersey Institute of Technology, Newark, New Jersey
| | - Ryan J Sullivan
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | - Genevieve M Boland
- Department of Surgery, Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | - Meenhard Herlyn
- Molecular and Cellular Oncogenesis Program and Melanoma Research Center, The Wistar Institute, Philadelphia, Pennsylvania
| | - Keith T Flaherty
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | - Nicola Zamboni
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Gao Zhang
- Molecular and Cellular Oncogenesis Program and Melanoma Research Center, The Wistar Institute, Philadelphia, Pennsylvania
| | | | - Wilhelm Krek
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Werner J Kovacs
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
23
|
Zou G, Bai J, Li D, Chen Y. Effect of metformin on the proliferation, apoptosis, invasion and autophagy of ovarian cancer cells. Exp Ther Med 2019; 18:2086-2094. [PMID: 31452705 PMCID: PMC6704536 DOI: 10.3892/etm.2019.7803] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 04/26/2019] [Indexed: 12/20/2022] Open
Abstract
The present study evaluated the effect of metformin on the SKOV3 ovarian cancer cell line and investigated the underlying mechanism. The inhibitory rate of SKOV3 cells was analyzed by MTT assay. SKOV3 cell apoptosis rate was quantitatively measured using flow cytometry. The effect of metformin on intracellular autophagosomes was observed using electron microscopy. The migration and invasion capabilities of SKOV3 cells were assessed by cell scratch test and Transwell assay. Results demonstrated that. the proliferation rate of SKOV3 cells was significantly inhibited in a time- and concentration-dependent manner following treatment with different concentrations of metformin for 24, 48 and 72 h. The number of migratory cells significantly decreased with increasing concentrations of metformin. The administration of metformin also promoted autophagy of ovarian cancer The expression level of microtubule associated protein 1 light chain 3-α protein was markedly upregulated. The mRNA expression level of metastasis-associated 1 (MTA1) was significantly downregulated following metformin treatment. In conclusion, metformin intervention suppressed SKOV3 proliferation and induced apoptosis in a concentration-dependent manner. Metformin also inhibited the invasion and migration of SKOV3 cells. It was hypothesized that the underlying mechanism of metformin's effect may involve MTA1 downregulation.
Collapse
Affiliation(s)
- Ge Zou
- Department of Obstetrics and Gynecology, North China University of Science and Technology Affiliated Hospital, Tangshan, Hebei 063000, P.R. China
| | - Jie Bai
- Department of Obstetrics and Gynecology, North China University of Science and Technology Affiliated Hospital, Tangshan, Hebei 063000, P.R. China
| | - Dandan Li
- Department of Obstetrics and Gynecology, North China University of Science and Technology Affiliated Hospital, Tangshan, Hebei 063000, P.R. China
| | - Yan Chen
- Department of Obstetrics and Gynecology, North China University of Science and Technology Affiliated Hospital, Tangshan, Hebei 063000, P.R. China
| |
Collapse
|
24
|
Li Y, Luo J, Lin MT, Zhi P, Guo WW, Han M, You J, Gao JQ. Co-Delivery of Metformin Enhances the Antimultidrug Resistant Tumor Effect of Doxorubicin by Improving Hypoxic Tumor Microenvironment. Mol Pharm 2019; 16:2966-2979. [PMID: 31095914 DOI: 10.1021/acs.molpharmaceut.9b00199] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Doxorubicin (DOX) is a first-line chemo drug for cancer therapy, yet it fails to treat multi-drug-resistant tumors. Hypoxia is a major causative factor leading to chemotherapy failure. Particularly, hypoxia up-regulates its responsive transcription factor-hypoxia-inducible factors (HIF)-to induce the overexpression of drug resistant genes. Metformin (MET) is recently found to cooperate with DOX against multiple tumors. As a mitochondrial inhibitor, MET could suppress tumor oxygen consumption, and thereby modulate the hypoxic tumor microenvironment. In this study, we used cationic liposomes to codeliver both DOX and MET for treating multi-drug-resistant breast cancer cells-MCF7/ADR. Faster release of MET enhanced the cytotoxicity of DOX through attenuating hypoxic stress both in vivo and in vitro. MET diminished the cellular oxygen consumption and inhibited HIF1α and P-glycoprotein (Pgp) expression in vitro. In addition, the dual-drug-loaded liposomes increased tumor targeting and intratumoral blood oxygen saturation, which suggested that the tumor reoxygenation effect of MET facilitated the exertion of its synergistic activity with DOX against MCF7/ADR xenografts. In general, our study represents a feasible strategy to boost the therapeutic effect in treating multi-drug-resistant cancer by improving the hypoxic tumor microenvironment.
Collapse
Affiliation(s)
- Ying Li
- Institute of Pharmaceutics, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou , People's Republic of China.,Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou , People's Republic of China
| | - Jing Luo
- Institute of Pharmaceutics, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou , People's Republic of China.,Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou , People's Republic of China
| | - Meng-Ting Lin
- Institute of Pharmaceutics, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou , People's Republic of China.,Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou , People's Republic of China
| | - Pei Zhi
- Institute of Pharmaceutics, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou , People's Republic of China.,Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou , People's Republic of China
| | - Wang-Wei Guo
- Institute of Pharmaceutics, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou , People's Republic of China.,Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou , People's Republic of China
| | - Min Han
- Institute of Pharmaceutics, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou , People's Republic of China.,Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou , People's Republic of China
| | - Jian You
- Institute of Pharmaceutics, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou , People's Republic of China.,Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou , People's Republic of China
| | - Jian-Qing Gao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou , People's Republic of China.,Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou , People's Republic of China
| |
Collapse
|
25
|
Inanc S, Keles D, Eskiizmir G, Basbinar Y, Oktay G. METFORMIN AND DICHOLOROACETATE COMBINATION EXERT A SYNERGISTIC EFFECT ON CELL VIABILITY OF ORAL SQUAMOUS CELL CARCINOMA. ENT UPDATES 2019. [DOI: 10.32448/entupdates.569464] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
26
|
Dichloroacetate and Salinomycin Exert a Synergistic Cytotoxic Effect in Colorectal Cancer Cell Lines. Sci Rep 2018; 8:17744. [PMID: 30531808 PMCID: PMC6288092 DOI: 10.1038/s41598-018-35815-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 11/07/2018] [Indexed: 12/18/2022] Open
Abstract
In the present study, we examined a hypothesis that dichloroacetate, a metabolic inhibitor, might efficiently potentiate the cytotoxic effect of salinomycin, an antibiotic ionophore, on two human colorectal cancer derived cell lines DLD-1 and HCT116. First, we performed a series of dose response experiments in the 2D cell culture by applying mono- and combination therapy and by using the Chou-Talalay method found that salinomycin in combination with dichloroacetate acted synergistically in both cell lines. Secondly, in order to recapitulate the in vivo tumor architecture, we tested various doses of these compounds, alone and in combination, in the 3D multicellular spheroid culture. The effect of combination of dichloracetate and salinomycin on multicellular spheroid size was stronger than the sum of both monotherapies, particularly in HCT116 cells. Further, we demonstrate that the synergistic effect of compounds may be related to the inhibitory effect of dichloroacetate on multidrug resistance proteins, and in contrast, it is not related to dichloroacetate-induced reduction of intracellular pH. Our findings indicate that the combination therapy of salinomycin and dichloroacetate could be an effective option for colorectal cancer treatment and provide the first mechanistic explanation of the synergistic action of these compounds.
Collapse
|
27
|
Florio R, De Lellis L, Veschi S, Verginelli F, di Giacomo V, Gallorini M, Perconti S, Sanna M, Mariani-Costantini R, Natale A, Arduini A, Amoroso R, Cataldi A, Cama A. Effects of dichloroacetate as single agent or in combination with GW6471 and metformin in paraganglioma cells. Sci Rep 2018; 8:13610. [PMID: 30206358 PMCID: PMC6134030 DOI: 10.1038/s41598-018-31797-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 08/06/2018] [Indexed: 12/14/2022] Open
Abstract
Paragangliomas (PGLs) are infiltrating autonomic nervous system tumors that cause important morbidity. At present, surgery is the only effective therapeutic option for this rare tumor. Thus, new agents for PGL treatment should be identified. Using unique PGL cell models established in our laboratory, we evaluated the effect of dichloroacetate (DCA) as single agent or in a novel combination with other metabolic drugs, including GW6471 and metformin. DCA and metformin had not been tested before in PGL. DCA reduced PGL cell viability and growth through mechanisms involving reactivation of PDH complex leading to promotion of oxidative metabolism, with lowering of lactate and enhanced ROS production. This resulted in cell cycle inhibition and induction of apoptosis in PGL cells, as shown by flow cytometry and immunoblot analyses. Moreover, DCA drastically impaired clonogenic activity and migration of PGL cells. Also metformin reduced PGL cell viability as single agent and the combinations of DCA, GW6471 and metformin had strong effects on cell viability. Furthermore, combined treatments had drastic and synergistic effects on clonogenic ability. In conclusion, DCA, GW6471 and metformin as single agents and in combination appear to have promising antitumor effects in unique cell models of PGL.
Collapse
Affiliation(s)
- Rosalba Florio
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy.,Unit of General Pathology, CeSI-MeT, University of Chieti, Chieti, Italy
| | - Laura De Lellis
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy. .,Unit of General Pathology, CeSI-MeT, University of Chieti, Chieti, Italy.
| | - Serena Veschi
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Fabio Verginelli
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy.,Unit of General Pathology, CeSI-MeT, University of Chieti, Chieti, Italy
| | - Viviana di Giacomo
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Marialucia Gallorini
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Silvia Perconti
- Unit of General Pathology, CeSI-MeT, University of Chieti, Chieti, Italy
| | - Mario Sanna
- Department of Otology and Skull Base Surgery, Gruppo Otologico, Piacenza, Italy
| | - Renato Mariani-Costantini
- Unit of General Pathology, CeSI-MeT, University of Chieti, Chieti, Italy.,Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Angelica Natale
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | | | - Rosa Amoroso
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Amelia Cataldi
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Alessandro Cama
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy. .,Unit of General Pathology, CeSI-MeT, University of Chieti, Chieti, Italy.
| |
Collapse
|
28
|
Zhang F, Chen H, Du J, Wang B, Yang L. Anticancer Activity of Metformin, an Antidiabetic Drug, Against Ovarian Cancer Cells Involves Inhibition of Cysteine-Rich 61 (Cyr61)/Akt/Mammalian Target of Rapamycin (mTOR) Signaling Pathway. Med Sci Monit 2018; 24:6093-6101. [PMID: 30171812 PMCID: PMC6130173 DOI: 10.12659/msm.909745] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Background Ovarian cancer is considered one of the lethal cancers responsible for high mortality and morbidity across the world. The prognosis and the survival rate of ovarian cancer is far from decent. Cysteine-rich 61 (Cyr61) also known as CCN1, is a member of CCN-family of growth factors, reported to be significantly overexpressed in several malignancies which include, but are not limited to, ovarian cancer. Recent studies have revealed that women with type 2 diabetes mellitus have an elevated risk of ovarian cancer. Hence, administration of an antidiabetic drug with anticancer effects such as metformin may act as an effective therapeutic regime against ovarian cancer. Material/Methods Cell viability and apoptosis were examined by MTT and Annexin V/PI double staining respectively. Cell migration was determined by Boyden Chamber assay. Transient knockdown of Cyr61 in ovarian cancer cells was achieved by transecting the cells with siRNA for Cyr61using Lipofectamine 2000. Results Our results indicated that treatment of ovarian cancer cells with metformin caused significant downregulation of Cyr61 protein expression levels ultimately favoring apoptosis. Transient knockdown of Cyr61 resulted in the inhibition of cell proliferation and migration. This was also associated with the concomitant downregulation of pAkt and pmTOR confirming the role of Cyr61 as an upstream modulator of Akt signaling. Conversely the extracellular supplementation of recombinant Cyr61 attenuates the cytotoxic properties of metformin in ovarian cancer cells. Conclusions Taken together, we concluded that metformin exhibits anticancer effects and Cyr61 acts as a direct target for metformin in ovarian cancer cells.
Collapse
Affiliation(s)
- Fengli Zhang
- Infertility Center, Qilu Hospital, Shandong University, Jinan, Shandong, China (mainland).,Department of Obstetrics and Gynecology, Liaocheng People's Hospital, Liaocheng, Shandong, China (mainland)
| | - Huixiao Chen
- Department of Obstetrics and Gynecology, Liaocheng People's Hospital, Liaocheng, Shandong, China (mainland)
| | - Jing Du
- Department of Obstetrics and Gynecology, Liaocheng People's Hospital, Liaocheng, Shandong, China (mainland)
| | - Bin Wang
- Department of Obstetrics and Gynecology, Liaocheng People's Hospital, Liaocheng, Shandong, China (mainland)
| | - Lixiao Yang
- Department of Obstetrics and Gynecology, Liaocheng People's Hospital, Liaocheng, Shandong, China (mainland)
| |
Collapse
|
29
|
Pan YH, Jiao L, Lin CY, Lu CH, Li L, Chen HY, Wang YB, He Y. Combined treatment with metformin and gefitinib overcomes primary resistance to EGFR-TKIs with EGFR mutation via targeting IGF-1R signaling pathway. Biologics 2018; 12:75-86. [PMID: 30154647 PMCID: PMC6108345 DOI: 10.2147/btt.s166867] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Aim Although EGFR tyrosine kinase inhibitors (TKIs) have shown dramatic effects against sensitizing EGFR mutations in non-small cell lung cancer (NSCLC), ~20%–30% of NSCLC patients with EGFR-sensitive mutation exhibit intrinsic resistance to EGFR-TKIs. The purpose of the current study was to investigate the enhanced antitumor effect of metformin (Met), a biguanide drug, in combination with gefitinib (Gef) in primary resistant human lung cancer cells and the associated molecular mechanism. Experimental design H1975 cell line was treated with Met and/or Gef to examine the inhibition of cell growth and potential mechanism of action by using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), Ki67 incorporation assay, flow cytometry analysis, small interfering RNA technology, Western blot analysis and xenograft implantation. Results Insulin-like growth factor-1 receptor (IGF-1R) signaling pathway was markedly activated in EGFR-TKI primary resistant H1975 cells as compared to EGFR-TKI acquired resistance cells (PC-9GR, H1650-M3) and EGFR-TKI sensitivity cells (PC-9, HCC827). Inhibition of IGF-1R activity by AG-1024 (a small molecule of IGF-1R inhibitor), as well as downregulation of IGF-1R by siRNA, significantly enhanced the ability of Gef to suppress proliferation and induce apoptosis in H1975 cells via the inhibition of AKT activation and subsequent upregulation of Bcl-2-interacting mediator of cell death (BIM). Interestingly, the observation showed that Met combined with Gef treatment had similar tumor growth suppression effects in comparison with the addition of AG-1024 to therapy with Gef. A clear synergistic antiproliferative interaction between Met and Gef was observed with a combination index (CI) value of 0.65. Notably, IGF-1R silencing mediated by RNA interference (RNAi) attenuated anticancer effects of Met without obviously resensitizing H1975 cells to Gef. Finally, Met-based combinatorial therapy effectively blocked tumor growth in the xenograft with TKI primary resistant lung cancer cells. Conclusion Our findings demonstrated that Met combined with Gef would be a promising strategy to overcome EGFR-TKI primary resistance via suppressing IGF-1R signaling pathway in NSCLC.
Collapse
Affiliation(s)
- Yong-Hong Pan
- Department of Respiratory Disease, Daping Hospital, Third Military Medical University, Chongqing 400042, China,
| | - Lin Jiao
- Department of Respiratory Disease, Daping Hospital, Third Military Medical University, Chongqing 400042, China,
| | - Cai-Yu Lin
- Department of Respiratory Disease, Daping Hospital, Third Military Medical University, Chongqing 400042, China,
| | - Cong-Hua Lu
- Department of Respiratory Disease, Daping Hospital, Third Military Medical University, Chongqing 400042, China,
| | - Li Li
- Department of Respiratory Disease, Daping Hospital, Third Military Medical University, Chongqing 400042, China,
| | - Heng-Yi Chen
- Department of Respiratory Disease, Daping Hospital, Third Military Medical University, Chongqing 400042, China,
| | - Yu-Bo Wang
- Department of Respiratory Disease, Daping Hospital, Third Military Medical University, Chongqing 400042, China,
| | - Yong He
- Department of Respiratory Disease, Daping Hospital, Third Military Medical University, Chongqing 400042, China,
| |
Collapse
|
30
|
Tavares‐Valente D, Granja S, Baltazar F, Queirós O. Bioenergetic modulators hamper cancer cell viability and enhance response to chemotherapy. J Cell Mol Med 2018; 22:3782-3794. [PMID: 29845734 PMCID: PMC6050502 DOI: 10.1111/jcmm.13642] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 03/14/2018] [Indexed: 01/12/2023] Open
Abstract
Gliomas are characterized by a marked glycolytic metabolism with a consequent production of massive amounts of lactate, even in the presence of normal levels of oxygen, associated to increased invasion capacity and to higher resistance to conventional treatment. This work aimed to understand how the metabolic modulation can influence tumour aggressive features and its potential to be used as complementary therapy. We assessed the effect of bioenergetic modulators (BMs) targeting different metabolic pathways in glioma cell characteristics. The in vivo effect of BMs was evaluated using the chicken chorioallantoic membrane model. Additionally, the effect of pre-treatment with BMs in the response to the antitumour drug temozolomide (TMZ) was analysed in vitro. Cell treatment with the BMs induced a decrease in cell viability and in migratory/invasion abilities, as well as modifications in metabolic parameters (glucose, lactate and ATP) and increased the cytotoxicity of the conventional drug TMZ. Furthermore, all BMs decreased the tumour growth and the number of blood vessels in an in vivo model. Our results demonstrate that metabolic modulation has the potential to be used as therapy to decrease the aggressiveness of the tumours or to be combined with conventional drugs used in glioma treatment.
Collapse
Affiliation(s)
- Diana Tavares‐Valente
- Life and Health Sciences Research Institute (ICVS)School of MedicineUniversity of MinhoCampus de Gualtar4710‐057BragaPortugal
- Department of SciencesIINFACTS ‐ Institute of Research and Advanced Training in Health Sciences and TechnologiesCESPU, CRLUniversity Institute of Health Sciences (IUCS)GandraPortugal
| | - Sara Granja
- Life and Health Sciences Research Institute (ICVS)School of MedicineUniversity of MinhoCampus de Gualtar4710‐057BragaPortugal
| | - Fátima Baltazar
- Life and Health Sciences Research Institute (ICVS)School of MedicineUniversity of MinhoCampus de Gualtar4710‐057BragaPortugal
| | - Odília Queirós
- Department of SciencesIINFACTS ‐ Institute of Research and Advanced Training in Health Sciences and TechnologiesCESPU, CRLUniversity Institute of Health Sciences (IUCS)GandraPortugal
| |
Collapse
|
31
|
Bai M, Yang L, Liao H, Liang X, Xie B, Xiong J, Tao X, Chen X, Cheng Y, Chen X, Feng Y, Zhang Z, Zheng W. Metformin sensitizes endometrial cancer cells to chemotherapy through IDH1-induced Nrf2 expression via an epigenetic mechanism. Oncogene 2018; 37:5666-5681. [PMID: 29921847 DOI: 10.1038/s41388-018-0360-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 05/19/2018] [Accepted: 05/25/2018] [Indexed: 02/07/2023]
Abstract
Chemoresistance is the major obstacle to cure endometrial cancer, whereas metformin has demonstrated sensitization to chemotherapy in endometrial cancer. A novel finding states that isocitrate dehydrogenase 1 (IDH1) involves in cancer chemoresistance. Recent studies have revealed that epigenetic modifications facilitate chemoresistance. However, whether IDH1 play a role in metformin-induced endometrial cancer chemosensitivity through epigenetic modification is incompletely understood. Immunohistochemistry and Elisa assays were used to evaluate the expression pattern of IDH1 in endometrial tissue and serum, respectively. Western blot was performed to determine changes in expression of key molecules in the IDH1-ɑ-KG-TET1-Nrf2 signaling pathway after various treatments. Dot blot assays were used to assess global hydroxymethylation levels after metformin administration or plasmid transfection. Antioxidant response element (ARE) activity in the IDH1 promoter region was monitored by luciferase assay. Cancer cell sensitivity to chemotherapy was detected by SRB assay. We found that activation of the IDH1 signaling pathway in endometrial cancer tissue resulting from aberrant expression of IDH1 and its downstream mediators conferred chemoresistance. We found that this effect was abated by metformin treatment. Dot blot and HMeDIP assays revealed that metformin blocked IDH1-ɑ-KG-TET1-mediated enhancement of Nrf2 hydroxymethylation levels, eliminating chemoresistance. Moreover, we observed that chemoresistance was enhanced via a regulatory loop in which Nrf2 activated IDH1-ɑ-KG-TET1-Nrf2 signaling via binding to the ARE sites in the IDH1 promoter region. Our findings highlight a critical role of IDH1-ɑ-KG-TET1-Nrf2 signaling in chemoresistance and suggest that rational combination therapy with metformin and chemotherapeutics has the potential to suppress chemoresistance.
Collapse
Affiliation(s)
- Mingzhu Bai
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, 200080, China
| | - Linlin Yang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, 200080, China.,Department of Obstetrics and Gynecology, Shanghai First People's Hospital, Baoshan Branch, Shanghai, 201900, China
| | - Hong Liao
- Department of Cervical Diseases, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200040, China
| | - Xiaoyan Liang
- Department of Obstetrics and Gynecology, Shanghai First People's Hospital, Baoshan Branch, Shanghai, 201900, China
| | - Bingying Xie
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, 200080, China
| | - Ji Xiong
- Department of Pathology, Huashan Hospital of Fudan University, Shanghai, 200040, China
| | - Xiang Tao
- Department of Pathology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China
| | - Xiong Chen
- Department of Obstetrics and Gynecology, Shanghai First People's Hospital, Baoshan Branch, Shanghai, 201900, China
| | - Yali Cheng
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China
| | - Xiaojun Chen
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China
| | - Youji Feng
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, 200080, China
| | - Zhenbo Zhang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, 200080, China. .,Department of Obstetrics and Gynecology, Shanghai First People's Hospital, Baoshan Branch, Shanghai, 201900, China.
| | - Wenxin Zheng
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA. .,Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
32
|
Han CY, Patten DA, Richardson RB, Harper ME, Tsang BK. Tumor metabolism regulating chemosensitivity in ovarian cancer. Genes Cancer 2018; 9:155-175. [PMID: 30603053 PMCID: PMC6305103 DOI: 10.18632/genesandcancer.176] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 08/14/2018] [Indexed: 12/26/2022] Open
Abstract
Elevated metabolism is a key hallmark of multiple cancers, serving to fulfill high anabolic demands. Ovarian cancer (OVCA) is the fifth leading cause of cancer deaths in women with a high mortality rate (45%). Chemoresistance is a major hurdle for OVCA treatment. Although substantial evidence suggests that metabolic reprogramming contributes to anti-apoptosis and the metastasis of multiple cancers, the link between tumor metabolism and chemoresistance in OVCA remains unknown. While clinical trials targeting metabolic reprogramming alone have been met with limited success, the synergistic effect of inhibiting tumor-specific metabolism with traditional chemotherapy warrants further examination, particularly in OVCA. This review summarizes the role of key glycolytic enzymes and other metabolic synthesis pathways in the progression of cancer and chemoresistance in OVCA. Within this context, mitochondrial dynamics (fission, fusion and cristae structure) are addressed regarding their roles in controlling metabolism and apoptosis, closely associated with chemosensitivity. The roles of multiple key oncogenes (Akt, HIF-1α) and tumor suppressors (p53, PTEN) in metabolic regulation are also described. Next, this review summarizes recent research of metabolism and future direction. Finally, we examine clinical drugs and inhibitors to target glycolytic metabolism, as well as the rationale for such strategies as potential therapeutics to overcome chemoresistant OVCA.
Collapse
Affiliation(s)
- Chae Young Han
- Department of Obstetrics and Gynecology and Cellular and Molecular Medicine, University of Ottawa, and Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - David A. Patten
- Canadian Nuclear Laboratories (CNL), Radiobiology and Health Branch, Chalk River Laboratories, Chalk River, Ontario, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Richard B. Richardson
- Canadian Nuclear Laboratories (CNL), Radiobiology and Health Branch, Chalk River Laboratories, Chalk River, Ontario, Canada
| | - Mary-Ellen Harper
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Benjamin K. Tsang
- Department of Obstetrics and Gynecology and Cellular and Molecular Medicine, University of Ottawa, and Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, China
| |
Collapse
|
33
|
HSF1 upregulates ATG4B expression and enhances epirubicin-induced protective autophagy in hepatocellular carcinoma cells. Cancer Lett 2017; 409:81-90. [DOI: 10.1016/j.canlet.2017.08.039] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Revised: 08/21/2017] [Accepted: 08/28/2017] [Indexed: 12/19/2022]
|
34
|
Han JE, Lim PW, Na CM, Choi YS, Lee JY, Kim Y, Park HW, Moon HE, Heo MS, Park HR, Kim DG, Paek SH. Inhibition of HIF1α and PDK Induces Cell Death of Glioblastoma Multiforme. Exp Neurobiol 2017; 26:295-306. [PMID: 29093638 PMCID: PMC5661062 DOI: 10.5607/en.2017.26.5.295] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 10/07/2017] [Accepted: 10/12/2017] [Indexed: 01/09/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive form of brain tumors. GBMs, like other tumors, rely relatively less on mitochondrial oxidative phosphorylation (OXPHOS) and utilize more aerobic glycolysis, and this metabolic shift becomes augmented under hypoxia. In the present study, we investigated the physiological significance of altered glucose metabolism and hypoxic adaptation in the GBM cell line U251 and two newly established primary GBMs (GBM28 and GBM37). We found that these three GBMs exhibited differential growth rates under hypoxia compared to those under normoxia. Under normoxia, the basal expressions of HIF1α and the glycolysis-associated genes, PDK1, PDK3, and GLUT1, were relatively low in U251 and GBM28, while their basal expressions were high in GBM37. Under hypoxia, the expressions of these genes were enhanced further in all three GBMs. Treatment with dichloroacetate (DCA), an inhibitor of pyruvate dehydrogenase kinase (PDK), induced cell death in GBM28 and GBM37 maintained under normoxia, whereas DCA effects disappeared under hypoxia, suggesting that hypoxic adaptation dominated DCA effects in these GBMs. In contrast, the inhibition of HIF1α with chrysin suppressed the expression of PDK1, PDK3, and GLUT1 and markedly promoted cell death of all GBMs under both normoxia and hypoxia. Interestingly, however, GBMs treated with chrysin under hypoxia still sustained higher viability than those under normoxia, and chrysin and DCA co-treatment was unable to eliminate this hypoxia-dependent resistance. Together, these results suggest that hypoxic adaptation is critical for maintaining viability of GBMs, and targeting hypoxic adaptation can be an important treatment option for GBMs.
Collapse
Affiliation(s)
- Jiwon Esther Han
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Pyung Won Lim
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Chul Min Na
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea
| | - You Sik Choi
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Joo Young Lee
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Yona Kim
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Hyung Woo Park
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Hyo Eun Moon
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Man Seung Heo
- Smart Healthcare Medical Device Research Center, Samsung Medical Center, Seoul 06351, Korea
| | - Hye Ran Park
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Dong Gyu Kim
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Sun Ha Paek
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul 03082, Korea.,Hypoxia Ischemia Disease Institute, Seoul National University College of Medicine, Seoul 03082, Korea
| |
Collapse
|
35
|
Li B, Li X, Xiong H, Zhou P, Ni Z, Yang T, Zhang Y, Zeng Y, He J, Yang F, Zhang N, Wang Y, Zheng Y, He F. Inhibition of COX2 enhances the chemosensitivity of dichloroacetate in cervical cancer cells. Oncotarget 2017; 8:51748-51757. [PMID: 28881683 PMCID: PMC5584284 DOI: 10.18632/oncotarget.18518] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 05/06/2017] [Indexed: 12/14/2022] Open
Abstract
Dichloroacetate (DCA), a traditional mitochondria-targeting agent, has shown promising prospect as a sensitizer in fighting against malignancies including cervical cancer. But it is unclear about the effect of DCA alone on cervical tumor. Moreover, previous reports have demonstrated that the increased cyclooxygenase-2 (COX2) expression is associated with chemoresistance and poor prognosis of cervical cancer. However, it is still unknown whether COX2 can affect the sensitivity of DCA in cervical cancer cells. In this study, we found that cervical cancer cells were insensitive to DCA. Furthermore, we for the first time revealed that DCA could upregulate COX2 which impeded the chemosensitivity of DCA in cervical cancer cells. Mechanistic study showed that DCA reduced the level of RNA binding protein quaking (QKI), leading to the decay suppression of COX2 mRNA and the subsequent elevation of COX2 protein. Inhibition of COX2 using celecoxib could sensitize DCA in repressing the growth of cervical cancer cells both in vitro and in vivo. These results indicate that COX2 is a novel resistance factor of DCA, and combination of celecoxib with DCA may be beneficial to the treatment of cervical cancer.
Collapse
Affiliation(s)
- Bo Li
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China
| | - Xinzhe Li
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China
| | - Haojun Xiong
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China
| | - Peng Zhou
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China
| | - Zhenhong Ni
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China
| | - Teng Yang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China
| | - Yan Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China
| | - Yijun Zeng
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China
| | - Jintao He
- Battalion 17 of Students, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Fan Yang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China
| | - Nan Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China
| | - Yuting Wang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China
| | - Yingru Zheng
- Department of Obstetrics and Gynecology, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing 400042, China
| | - Fengtian He
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China
| |
Collapse
|