1
|
Liaghat M, Ferdousmakan S, Mortazavi SH, Yahyazadeh S, Irani A, Banihashemi S, Seyedi Asl FS, Akbari A, Farzam F, Aziziyan F, Bakhtiyari M, Arghavani MJ, Zalpoor H, Nabi-Afjadi M. The impact of epithelial-mesenchymal transition (EMT) induced by metabolic processes and intracellular signaling pathways on chemo-resistance, metastasis, and recurrence in solid tumors. Cell Commun Signal 2024; 22:575. [PMID: 39623377 PMCID: PMC11610171 DOI: 10.1186/s12964-024-01957-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 11/22/2024] [Indexed: 12/06/2024] Open
Abstract
The intricate cellular process, known as the epithelial-mesenchymal transition (EMT), significantly influences solid tumors development. Changes in cell shape, metabolism, and gene expression linked to EMT facilitate tumor cell invasion, metastasis, drug resistance, and recurrence. So, a better understanding of the intricate processes underlying EMT and its role in tumor growth may lead to the development of novel therapeutic approaches for the treatment of solid tumors. This review article focuses on the signals that promote EMT and metabolism, the intracellular signaling pathways leading to EMT, and the network of interactions between EMT and cancer cell metabolism. Furthermore, the functions of EMT in treatment resistance, recurrence, and metastasis of solid cancers are covered. Lastly, treatment approaches that focus on intracellular signaling networks and metabolic alterations brought on by EMT will be discussed.
Collapse
Affiliation(s)
- Mahsa Liaghat
- Department of Medical Laboratory Sciences, Faculty of Medical Sciences, Kazerun Branch, Islamic Azad University, Kazerun, Iran
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Saeid Ferdousmakan
- Department of Pharmacy Practice, Nargund College of Pharmacy, Bangalore, 560085, India
| | | | - Sheida Yahyazadeh
- Department of Immunology, Faculty of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Asrin Irani
- Department of Biology, Faculty of Basic Sciences, University of Guilan, Rasht, Iran
| | - Sara Banihashemi
- Department of Bioscience, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | | | - Abdullatif Akbari
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farnoosh Farzam
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Aziziyan
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Maryam Bakhtiyari
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
- Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Mohammad Javad Arghavani
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Hamidreza Zalpoor
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran.
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
2
|
Liu WJ, Wang L, Sun FL, Zhou FM, Zhang RK, Liu J, Zhao M, Wang LH, Qin YR, Zhao YQ, Qiu JG, Jiang BH. Hexavalent chromium induced metabolic reprogramming, carcinogenesis and tumor progression through PDK1 upregulation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 288:117341. [PMID: 39550876 DOI: 10.1016/j.ecoenv.2024.117341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/06/2024] [Accepted: 11/10/2024] [Indexed: 11/19/2024]
Abstract
Lung cancer is the leading factor of cancer-related death in the worldwide. Hexavalent chromium [Cr(VI)] is a potential carcinogen for inducing lung cancers. To understand new mechanism of Cr(VI)-induced tumorigenesis and cancer development, we identified that PDK1 expression levels were greatly increased in chromium-transformed cells (Cr-T) compared to the parental BEAS-2B (B2B) cells by proteomic profiling and Western blotting; PDK1 levels were also induced in lung cancer cell lines and in lung samples of mice exposed to Cr(VI). Cr(VI) increased Warburg effect, cell migration, proliferation and colony formation through PDK1 upregulation. To identify the mechanism of PDK1 induction, we performed miRNA-seq analysis of Cr-T and B2B cells, and found miR-493 levels was significantly suppressed by Cr(VI). PDK1 was induced by miR-493 suppression, and was a direct target of miR-493. Interestingly, we also found HIF-1α was directly targeting by miR-493 and was induced by miR-493 downregulation. HIF-1α expression levels were upregulated in lung samples of mice with Cr(VI)-exposure. PDK1 was induced by HIF-1α, showing miR-493 suppression can directly induce PDK1 as well as through HIF-1α induction. MiR-493 overexpression was sufficient to suppress tumor growth, PDK1 and HIF-1α expression in vivo. We also showed that levels of miR-493 suppression, HIF-1α and PDK1 elevations were strongly correlated with poor prognosis of lung cancer subjects. These results demonstrate both HIF-1α and PDK1 expression are induced by Cr(VI)-mediated miR-493 suppression, and MiR-493/HIF-1α/PDK1 axis is a new pathway in Cr(VI)-inducing carcinogenesis and tumor growth.
Collapse
Affiliation(s)
- Wen-Jing Liu
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Lin Wang
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China; Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Fan-Li Sun
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Feng-Mei Zhou
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Rui-Ke Zhang
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Jie Liu
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Min Zhao
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Li-Hong Wang
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Yan-Ru Qin
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan China
| | - Yan-Qiu Zhao
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Jian-Ge Qiu
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China; Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan 450000, China.
| | - Bing-Hua Jiang
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China; Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan 450000, China.
| |
Collapse
|
3
|
Ning Z, Liu K, Xiong H. CD39 is induced by nutrient deprivation in colorectal cancer cells and contributes to the tolerance to hypoxia. Biochem Biophys Res Commun 2024; 730:150367. [PMID: 38991255 DOI: 10.1016/j.bbrc.2024.150367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 07/04/2024] [Accepted: 07/05/2024] [Indexed: 07/13/2024]
Abstract
Rapid tumor growth and insufficient blood supply leads to the development of a hypoxic and nutrient deprived microenvironment. To survive, tumor cells need to tolerate these adverse conditions. Here we found the expression of CD39 was enhanced in necrotic regions distant from blood vessels. We speculate that this is a strategy for tumor cells to actively adapt to the hostile environment. Further studies showed that CD39 was induced by nutrient deprivation through the AMPK signalling pathway. We next explored the significance of CD39 for tumor cells. Our results showed that CD39 reduced cellular oxygen consumption, which could be significant for tumor cells if the available oxygen is limited. Metabolomics analysis showed that overexpression of CD39 significantly altered cellular metabolism, and tricarboxylic acid (TCA) cycle was identified as the most impacted metabolic pathway. In order to explore the molecular mechanism, we performed RNA-seq analysis. The results showed that CD39 significantly up-regulated the expression of pyruvate dehydrogenase kinase isozyme 2 (PDK2), thus inhibiting the activity of pyruvate dehydrogenase (PDH) and TCA cycle. Finally, CD39 was shown to protect tumor cells from hypoxia-induced cell death and reduce intratumoral hypoxia levels. CD39 has attracted a great deal of attention as a newly discovered immune checkpoint molecule in recent years. Our results indicate that CD39 not only plays a role in immune regulation, but also enables tumor cells to tolerate hypoxia by inhibiting TCA cycle and reducing cellular oxygen consumption. This study provides evidence that targeting CD39 may be a novel strategy to prevent adaptation of tumor cells in stressed conditions.
Collapse
Affiliation(s)
- Zhaochen Ning
- Jining Key Laboratory of Immunology, Jining Medical University, Jining, 272067, China
| | - Keyan Liu
- Department of Public Health, Jining Medical University, Jining, 272067, China
| | - Huabao Xiong
- Jining Key Laboratory of Immunology, Jining Medical University, Jining, 272067, China.
| |
Collapse
|
4
|
Zhang H, Liu X, Li J, Meng J, Huang W, Su X, Zhang X, Gao G, Wang X, Su H, Zhang F, Zhang T. ING5 inhibits aerobic glycolysis of lung cancer cells by promoting TIE1-mediated phosphorylation of pyruvate dehydrogenase kinase 1 at Y163. Front Med 2024; 18:878-895. [PMID: 39269568 DOI: 10.1007/s11684-024-1057-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 12/04/2023] [Indexed: 09/15/2024]
Abstract
Aerobic glycolysis is critical for tumor growth and metastasis. Previously, we have found that the overexpression of the inhibitor of growth 5 (ING5) inhibits lung cancer aggressiveness and epithelial-mesenchymal transition (EMT). However, whether ING5 regulates lung cancer metabolism reprogramming remains unknown. Here, by quantitative proteomics, we showed that ING5 differentially regulates protein phosphorylation and identified a new site (Y163) of the key glycolytic enzyme PDK1 whose phosphorylation was upregulated 13.847-fold. By clinical study, decreased p-PDK1Y163 was observed in lung cancer tissues and correlated with poor survival. p-PDK1Y163 represents the negative regulatory mechanism of PDK1 by causing PDHA1 dephosphorylation and activation, leading to switching from glycolysis to oxidative phosphorylation, with increasing oxygen consumption and decreasing lactate production. These effects could be impaired by PDK1Y163F mutation, which also impaired the inhibitory effects of ING5 on cancer cell EMT and invasiveness. Mouse xenograft models confirmed the indispensable role of p-PDK1Y163 in ING5-inhibited tumor growth and metastasis. By siRNA screening, ING5-upregulated TIE1 was identified as the upstream tyrosine protein kinase targeting PDK1Y163. TIE1 knockdown induced the dephosphorylation of PDK1Y163 and increased the migration and invasion of lung cancer cells. Collectively, ING5 overexpression-upregulated TIE1 phosphorylates PDK1Y163, which is critical for the inhibition of aerobic glycolysis and invasiveness of lung cancer cells.
Collapse
Affiliation(s)
- Haihua Zhang
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Xinli Liu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710038, China
| | - Junqiang Li
- Department of Oncology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Jin Meng
- Department of Pharmacy, the Medical Security Centre, Chinese PLA General Hospital, Beijing, 100091, China
| | - Wan Huang
- National Translational Science Center for Molecular Medicine and Department of Cell Biology, Fourth Military Medical University, Xi'an, 710038, China
| | - Xuan Su
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Xutao Zhang
- Aerospace Clinical Medical Center, School of Aerospace Medicine, Fourth Military Medical University, Xi'an, 710038, China
| | - Guizhou Gao
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Xiaodong Wang
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Haichuan Su
- Department of Oncology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China.
| | - Feng Zhang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710038, China.
| | - Tao Zhang
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China.
| |
Collapse
|
5
|
Pu J, Han J, Yang J, Yu L, Wan H. Anaerobic Glycolysis and Ischemic Stroke: From Mechanisms and Signaling Pathways to Natural Product Therapy. ACS Chem Neurosci 2024; 15:3090-3105. [PMID: 39140296 DOI: 10.1021/acschemneuro.4c00371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024] Open
Abstract
Ischemic stroke is a serious condition that results in high rates of illness and death. Anaerobic glycolysis becomes the primary means of providing energy to the brain during periods of low oxygen levels, such as in the aftermath of an ischemic stroke. This process is essential for maintaining vital brain functions and has significant implications for recovery following a stroke. Energy supply by anaerobic glycolysis and acidosis caused by lactic acid accumulation are important pathological processes after ischemic stroke. Numerous natural products regulate glucose and lactate, which in turn modulate anaerobic glycolysis. This article focuses on the relationship between anaerobic glycolysis and ischemic stroke, as well as the associated signaling pathways and natural products that play a therapeutic role. These natural products, which can regulate anaerobic glycolysis, will provide new avenues and perspectives for the treatment of ischemic stroke in the future.
Collapse
Affiliation(s)
- Jia Pu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jin Han
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jiehong Yang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Li Yu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Center of Safety Evaluation and Research, Hangzhou Medical College, Hangzhou, Zhejiang 310053, China
| | - Haitong Wan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| |
Collapse
|
6
|
Nishitsuji K, Mito R, Ikezaki M, Yano H, Fujiwara Y, Matsubara E, Nishikawa T, Ihara Y, Uchimura K, Iwahashi N, Sakagami T, Suzuki M, Komohara Y. Impacts of cytoplasmic p53 aggregates on the prognosis and the transcriptome in lung squamous cell carcinoma. Cancer Sci 2024; 115:2947-2960. [PMID: 39031627 PMCID: PMC11462941 DOI: 10.1111/cas.16252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 07/22/2024] Open
Abstract
The tumor suppressor TP53 gene, the most frequently mutated gene in human cancers, produces the product tumor protein p53, which plays an essential role in DNA damage. p53 protein mutations may contribute to tumorigenesis by loss of tumor suppressive functions and malignancy of cancer cells via gain-of-oncogenic functions. We previously reported that mutant p53 proteins form aggregates and that cytoplasmic p53 aggregates were associated with poor prognosis in human ovarian cancer. However, the prognostic impact of p53 aggregation in other tumors including lung squamous cell carcinoma (SCC) is poorly understood. Here, we demonstrated that lung SCC cases with cytoplasmic p53 aggregates had a significantly poor clinical prognosis. Analysis via patient-derived tumor organoids (PDOs) established from lung SCC patients and possessing cytoplasmic p53 aggregates showed that eliminating cytoplasmic p53 aggregates suppressed cell proliferation. RNA sequencing and transcriptome analysis of p53 aggregate-harboring PDOs indicated multiple candidate pathways involved in p53 aggregate oncogenic functions. With lung SCC-derived cell lines, we found that cytoplasmic p53 aggregates contributed to cisplatin resistance. This study thus shows that p53 aggregates are a predictor of poor prognosis in lung SCC and suggests that detecting p53 aggregates via p53 conventional immunohistochemical analysis may aid patient selection for platinum-based therapy.
Collapse
Affiliation(s)
- Kazuchika Nishitsuji
- Department of Biochemistry, School of MedicineWakayama Medical UniversityWakayamaJapan
- Unité de Glycobiologie Structurale et FonctionnelleUMR 8576 CNRS, Université de LilleVilleneuve d'AscqFrance
| | - Remi Mito
- Department of Cell Pathology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
- Department of Respiratory Medicine, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Midori Ikezaki
- Department of Biochemistry, School of MedicineWakayama Medical UniversityWakayamaJapan
| | - Hiromu Yano
- Department of Cell Pathology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Yukio Fujiwara
- Department of Cell Pathology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Eri Matsubara
- Department of Cell Pathology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
- Department of Thoracic and Breast Surgery, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Taro Nishikawa
- Department of Biochemistry, School of MedicineWakayama Medical UniversityWakayamaJapan
| | - Yoshito Ihara
- Department of Biochemistry, School of MedicineWakayama Medical UniversityWakayamaJapan
| | - Kenji Uchimura
- Unité de Glycobiologie Structurale et FonctionnelleUMR 8576 CNRS, Université de LilleVilleneuve d'AscqFrance
| | - Naoyuki Iwahashi
- Department of Obstetrics and Gynecology, School of MedicineWakayama Medical UniversityWakayamaJapan
| | - Takuro Sakagami
- Department of Respiratory Medicine, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Makoto Suzuki
- Department of Thoracic and Breast Surgery, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Yoshihiro Komohara
- Department of Cell Pathology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
- Center for Metabolic Regulation of Healthy AgingKumamoto UniversityKumamotoJapan
| |
Collapse
|
7
|
Mailloux RJ. The emerging importance of the α-keto acid dehydrogenase complexes in serving as intracellular and intercellular signaling platforms for the regulation of metabolism. Redox Biol 2024; 72:103155. [PMID: 38615490 PMCID: PMC11021975 DOI: 10.1016/j.redox.2024.103155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/04/2024] [Accepted: 04/09/2024] [Indexed: 04/16/2024] Open
Abstract
The α-keto acid dehydrogenase complex (KDHc) class of mitochondrial enzymes is composed of four members: pyruvate dehydrogenase (PDHc), α-ketoglutarate dehydrogenase (KGDHc), branched-chain keto acid dehydrogenase (BCKDHc), and 2-oxoadipate dehydrogenase (OADHc). These enzyme complexes occupy critical metabolic intersections that connect monosaccharide, amino acid, and fatty acid metabolism to Krebs cycle flux and oxidative phosphorylation (OxPhos). This feature also imbues KDHc enzymes with the heightened capacity to serve as platforms for propagation of intracellular and intercellular signaling. KDHc enzymes serve as a source and sink for mitochondrial hydrogen peroxide (mtH2O2), a vital second messenger used to trigger oxidative eustress pathways. Notably, deactivation of KDHc enzymes through reversible oxidation by mtH2O2 and other electrophiles modulates the availability of several Krebs cycle intermediates and related metabolites which serve as powerful intracellular and intercellular messengers. The KDHc enzymes also play important roles in the modulation of mitochondrial metabolism and epigenetic programming in the nucleus through the provision of various acyl-CoAs, which are used to acylate proteinaceous lysine residues. Intriguingly, nucleosomal control by acylation is also achieved through PDHc and KGDHc localization to the nuclear lumen. In this review, I discuss emerging concepts in the signaling roles fulfilled by the KDHc complexes. I highlight their vital function in serving as mitochondrial redox sensors and how this function can be used by cells to regulate the availability of critical metabolites required in cell signaling. Coupled with this, I describe in detail how defects in KDHc function can cause disease states through the disruption of cell redox homeodynamics and the deregulation of metabolic signaling. Finally, I propose that the intracellular and intercellular signaling functions of the KDHc enzymes are controlled through the reversible redox modification of the vicinal lipoic acid thiols in the E2 subunit of the complexes.
Collapse
Affiliation(s)
- Ryan J Mailloux
- School of Human Nutrition, Faculty of Agricultural and Environmental Sciences, McGill University, Ste-Anne-de-Bellevue, Quebec, Canada.
| |
Collapse
|
8
|
Zhao J, Ma X, Gao P, Han X, Zhao P, Xie F, Liu M. Advancing glioblastoma treatment by targeting metabolism. Neoplasia 2024; 51:100985. [PMID: 38479191 PMCID: PMC10950892 DOI: 10.1016/j.neo.2024.100985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 03/04/2024] [Indexed: 03/24/2024]
Abstract
Alterations in cellular metabolism are important hallmarks of glioblastoma(GBM). Metabolic reprogramming is a critical feature as it meets the higher nutritional demand of tumor cells, including proliferation, growth, and survival. Many genes, proteins, and metabolites associated with GBM metabolism reprogramming have been found to be aberrantly expressed, which may provide potential targets for cancer treatment. Therefore, it is becoming increasingly important to explore the role of internal and external factors in metabolic regulation in order to identify more precise therapeutic targets and diagnostic markers for GBM. In this review, we define the metabolic characteristics of GBM, investigate metabolic specificities such as targetable vulnerabilities and therapeutic resistance, as well as present current efforts to target GBM metabolism to improve the standard of care.
Collapse
Affiliation(s)
- Jinyi Zhao
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China; Beijing Molecular Hydrogen Research Center, Beijing, China
| | - Xuemei Ma
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China; Beijing Molecular Hydrogen Research Center, Beijing, China
| | - Peixian Gao
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China; Beijing Molecular Hydrogen Research Center, Beijing, China
| | - Xueqi Han
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China; Beijing Molecular Hydrogen Research Center, Beijing, China
| | - Pengxiang Zhao
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China; Beijing Molecular Hydrogen Research Center, Beijing, China
| | - Fei Xie
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China; Beijing Molecular Hydrogen Research Center, Beijing, China
| | - Mengyu Liu
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China; Beijing Molecular Hydrogen Research Center, Beijing, China.
| |
Collapse
|
9
|
Liu X, Li J, Huang Q, Jin M, Huang G. Ginsenoside Rh2 shifts tumor metabolism from aerobic glycolysis to oxidative phosphorylation through regulating the HIF1-α/PDK4 axis in non-small cell lung cancer. Mol Med 2024; 30:56. [PMID: 38671369 PMCID: PMC11055298 DOI: 10.1186/s10020-024-00813-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 03/18/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Ginsenoside Rh2 (G-Rh2), a steroidal compound extracted from roots of ginseng, has been extensively studied in tumor therapy. However, its specific regulatory mechanism in non-small cell lung cancer (NSCLC) is not well understood. Pyruvate dehydrogenase kinase 4 (PDK4), a central regulator of cellular energy metabolism, is highly expressed in various malignant tumors. We investigated the impact of G-Rh2 on the malignant progression of NSCLC and how it regulated PDK4 to influence tumor aerobic glycolysis and mitochondrial function. METHOD We examined the inhibitory effect of G-Rh2 on NSCLC through I proliferation assay, migration assay and flow cytometry in vitro. Subsequently, we verified the ability of G-Rh2 to inhibit tumor growth and metastasis by constructing subcutaneous tumor and metastasis models in nude mice. Proteomics analysis was conducted to analyze the action pathways of G-Rh2. Additionally, we assessed glycolysis and mitochondrial function using seahorse, PET-CT, Western blot, and RT-qPCR. RESULT Treatment with G-Rh2 significantly inhibited tumor proliferation and migration ability both in vitro and in vivo. Furthermore, G-Rh2 inhibited the tumor's aerobic glycolytic capacity, including glucose uptake and lactate production, through the HIF1-α/PDK4 pathway. Overexpression of PDK4 demonstrated that G-Rh2 targeted the inhibition of PDK4 expression, thereby restoring mitochondrial function, promoting reactive oxygen species (ROS) accumulation, and inducing apoptosis. When combined with sodium dichloroacetate, a PDK inhibitor, it complemented the inhibitory capacity of PDKs, acting synergistically as a detoxifier. CONCLUSION G-Rh2 could target and down-regulate the expression of HIF-1α, resulting in decreased expression of glycolytic enzymes and inhibition of aerobic glycolysis in tumors. Additionally, by directly targeting mitochondrial PDK, it elevated mitochondrial oxidative phosphorylation and enhanced ROS accumulation, thereby promoting tumor cells to undergo normal apoptotic processes.
Collapse
Affiliation(s)
- Xiyu Liu
- Shanghai University of Traditional Chinese Medicine, 201203, Shanghai, P.R. China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Road, Pudong New Area, 201318, Shanghai, China
| | - Jingjing Li
- Shanghai University of Traditional Chinese Medicine, 201203, Shanghai, P.R. China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Road, Pudong New Area, 201318, Shanghai, China
| | - Qingqing Huang
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Road, Pudong New Area, 201318, Shanghai, China.
| | - Mingming Jin
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Road, Pudong New Area, 201318, Shanghai, China.
| | - Gang Huang
- Shanghai University of Traditional Chinese Medicine, 201203, Shanghai, P.R. China.
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Road, Pudong New Area, 201318, Shanghai, China.
| |
Collapse
|
10
|
Toniyan KA, Malkov AA, Biryukov NS, Gorbacheva EY, Boyarintsev VV, Ogneva IV. The Cellular Respiration of Endometrial Biopsies from Patients with Various Forms of Endometriosis. Int J Mol Sci 2024; 25:3680. [PMID: 38612490 PMCID: PMC11011257 DOI: 10.3390/ijms25073680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/19/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
Endometriosis is one of the leading pathologies of the reproductive system of women of fertile age, which shows changes in cell metabolism in the lesions. We conducted a study of the cellular respiration according to the polarography and the mRNA content of the main metabolic proteins using qRT-PCR of intraoperative endometrial biopsies from patients in the control group and with different localizations of endometriosis (adenomyosis, endometrioma, pelvic peritoneum). In biopsy samples of patients with endometriomas and pelvic peritoneum endometriotic lesions, the rate of oxygen absorption was significantly reduced, and, moreover, in the extragenital case, there was a shift to succinate utilization. The mRNA content of the cytochrome c, cytochrome c oxidase, and ATP synthase was also reduced, but hexokinase HK2 as well as pyruvate kinase were significantly higher than in the control. These oxidative phosphorylation and gene expression profiles suggest the Warburg effect and a shift in metabolism toward glycolysis. For adenomyosis, on the contrary, cellular respiration was significantly higher than in the control group due to the terminal region of the respiratory chain, ATP synthase, and its mRNA was increased as well. These data allow us to suggest that the therapeutic strategies of endometriosis based on modulation energy metabolism should take lesion localization into account.
Collapse
Affiliation(s)
- Konstantin A. Toniyan
- Cell Biophysics Laboratory, State Scientific Center of the Russian Federation Institute of Biomedical Problems of the Russian Academy of Sciences, 123007 Moscow, Russia; (K.A.T.); (A.A.M.); (N.S.B.); (E.Y.G.)
- Gynecology Department, FGBU KB1 (Volynskaya) UDP RF, 121352 Moscow, Russia
| | - Artyom A. Malkov
- Cell Biophysics Laboratory, State Scientific Center of the Russian Federation Institute of Biomedical Problems of the Russian Academy of Sciences, 123007 Moscow, Russia; (K.A.T.); (A.A.M.); (N.S.B.); (E.Y.G.)
- Medical and Biological Physics Department, I.M. Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya Street, 119991 Moscow, Russia
| | - Nikolay S. Biryukov
- Cell Biophysics Laboratory, State Scientific Center of the Russian Federation Institute of Biomedical Problems of the Russian Academy of Sciences, 123007 Moscow, Russia; (K.A.T.); (A.A.M.); (N.S.B.); (E.Y.G.)
- Medical and Biological Physics Department, I.M. Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya Street, 119991 Moscow, Russia
| | - Elena Yu. Gorbacheva
- Cell Biophysics Laboratory, State Scientific Center of the Russian Federation Institute of Biomedical Problems of the Russian Academy of Sciences, 123007 Moscow, Russia; (K.A.T.); (A.A.M.); (N.S.B.); (E.Y.G.)
- Gynecology Department, FGBU KB1 (Volynskaya) UDP RF, 121352 Moscow, Russia
| | - Valery V. Boyarintsev
- Emergency and Extreme Medicine Department, FGBU DPO CGMA UDP RF, 121359 Moscow, Russia;
| | - Irina V. Ogneva
- Cell Biophysics Laboratory, State Scientific Center of the Russian Federation Institute of Biomedical Problems of the Russian Academy of Sciences, 123007 Moscow, Russia; (K.A.T.); (A.A.M.); (N.S.B.); (E.Y.G.)
- Medical and Biological Physics Department, I.M. Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya Street, 119991 Moscow, Russia
| |
Collapse
|
11
|
Petralia F, Ma W, Yaron TM, Caruso FP, Tignor N, Wang JM, Charytonowicz D, Johnson JL, Huntsman EM, Marino GB, Calinawan A, Evangelista JE, Selvan ME, Chowdhury S, Rykunov D, Krek A, Song X, Turhan B, Christianson KE, Lewis DA, Deng EZ, Clarke DJB, Whiteaker JR, Kennedy JJ, Zhao L, Segura RL, Batra H, Raso MG, Parra ER, Soundararajan R, Tang X, Li Y, Yi X, Satpathy S, Wang Y, Wiznerowicz M, González-Robles TJ, Iavarone A, Gosline SJC, Reva B, Robles AI, Nesvizhskii AI, Mani DR, Gillette MA, Klein RJ, Cieslik M, Zhang B, Paulovich AG, Sebra R, Gümüş ZH, Hostetter G, Fenyö D, Omenn GS, Cantley LC, Ma'ayan A, Lazar AJ, Ceccarelli M, Wang P. Pan-cancer proteogenomics characterization of tumor immunity. Cell 2024; 187:1255-1277.e27. [PMID: 38359819 PMCID: PMC10988632 DOI: 10.1016/j.cell.2024.01.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 09/29/2023] [Accepted: 01/16/2024] [Indexed: 02/17/2024]
Abstract
Despite the successes of immunotherapy in cancer treatment over recent decades, less than <10%-20% cancer cases have demonstrated durable responses from immune checkpoint blockade. To enhance the efficacy of immunotherapies, combination therapies suppressing multiple immune evasion mechanisms are increasingly contemplated. To better understand immune cell surveillance and diverse immune evasion responses in tumor tissues, we comprehensively characterized the immune landscape of more than 1,000 tumors across ten different cancers using CPTAC pan-cancer proteogenomic data. We identified seven distinct immune subtypes based on integrative learning of cell type compositions and pathway activities. We then thoroughly categorized unique genomic, epigenetic, transcriptomic, and proteomic changes associated with each subtype. Further leveraging the deep phosphoproteomic data, we studied kinase activities in different immune subtypes, which revealed potential subtype-specific therapeutic targets. Insights from this work will facilitate the development of future immunotherapy strategies and enhance precision targeting with existing agents.
Collapse
Affiliation(s)
- Francesca Petralia
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Weiping Ma
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Tomer M Yaron
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA; Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA; Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Francesca Pia Caruso
- BIOGEM Institute of Molecular Biology and Genetics, 83031 Ariano Irpino, Italy; Department of Electrical Engineering and Information Technologies, University of Naples "Federico II", Naples, Italy
| | - Nicole Tignor
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Joshua M Wang
- Institute for Systems Genetics, New York University Grossman School of Medicine, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Daniel Charytonowicz
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jared L Johnson
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Emily M Huntsman
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA; Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Giacomo B Marino
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Anna Calinawan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - John Erol Evangelista
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Myvizhi Esai Selvan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Shrabanti Chowdhury
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Dmitry Rykunov
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Azra Krek
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Xiaoyu Song
- Institute for Healthcare Delivery Science, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Berk Turhan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Karen E Christianson
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - David A Lewis
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Eden Z Deng
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Daniel J B Clarke
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jeffrey R Whiteaker
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Jacob J Kennedy
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Lei Zhao
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Rossana Lazcano Segura
- Departments of Pathology & Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Harsh Batra
- Department of Translational Molecular Pathology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Maria Gabriela Raso
- Department of Translational Molecular Pathology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Edwin Roger Parra
- Department of Translational Molecular Pathology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Rama Soundararajan
- Department of Translational Molecular Pathology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ximing Tang
- Department of Translational Molecular Pathology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yize Li
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Xinpei Yi
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Shankha Satpathy
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Ying Wang
- Institute for Systems Genetics, New York University Grossman School of Medicine, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Maciej Wiznerowicz
- Department of Medical Biotechnology, Poznan University of Medical Sciences, 61-701 Poznań, Poland; International Institute for Molecular Oncology, 60-203 Poznań, Poland; Department of Oncology, Heliodor Swiecicki Clinical Hospital, 60-203 Poznań, Poland
| | - Tania J González-Robles
- Institute for Systems Genetics, New York University Grossman School of Medicine, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Antonio Iavarone
- Department of Neurological Surgery, Department of Biochemistry, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Sara J C Gosline
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Boris Reva
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ana I Robles
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, Rockville, MD 20850, USA
| | - Alexey I Nesvizhskii
- Departments of Pathology and Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - D R Mani
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Michael A Gillette
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA; Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Robert J Klein
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Marcin Cieslik
- Departments of Pathology and Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Bing Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Amanda G Paulovich
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Robert Sebra
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zeynep H Gümüş
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Galen Hostetter
- Pathology and Biorepository Core, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - David Fenyö
- Institute for Systems Genetics, New York University Grossman School of Medicine, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Gilbert S Omenn
- Departments of Computational Medicine & Bioinformatics, Internal Medicine, Human Genetics, & Environmental Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lewis C Cantley
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Avi Ma'ayan
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Alexander J Lazar
- Departments of Pathology & Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michele Ceccarelli
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA; Department of Public Health Sciences, University of Miami, Miami, FL, USA
| | - Pei Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
12
|
Pucci G, Minafra L, Bravatà V, Calvaruso M, Turturici G, Cammarata FP, Savoca G, Abbate B, Russo G, Cavalieri V, Forte GI. Glut-3 Gene Knockdown as a Potential Strategy to Overcome Glioblastoma Radioresistance. Int J Mol Sci 2024; 25:2079. [PMID: 38396757 PMCID: PMC10889562 DOI: 10.3390/ijms25042079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
The hypoxic pattern of glioblastoma (GBM) is known to be a primary cause of radioresistance. Our study explored the possibility of using gene knockdown of key factors involved in the molecular response to hypoxia, to overcome GBM radioresistance. We used the U87 cell line subjected to chemical hypoxia generated by CoCl2 and exposed to 2 Gy of X-rays, as single or combined treatments, and evaluated gene expression changes of biomarkers involved in the Warburg effect, cell cycle control, and survival to identify the best molecular targets to be knocked-down, among those directly activated by the HIF-1α transcription factor. By this approach, glut-3 and pdk-1 genes were chosen, and the effects of their morpholino-induced gene silencing were evaluated by exploring the proliferative rates and the molecular modifications of the above-mentioned biomarkers. We found that, after combined treatments, glut-3 gene knockdown induced a greater decrease in cell proliferation, compared to pdk-1 gene knockdown and strong upregulation of glut-1 and ldha, as a sign of cell response to restore the anaerobic glycolysis pathway. Overall, glut-3 gene knockdown offered a better chance of controlling the anaerobic use of pyruvate and a better proliferation rate reduction, suggesting it is a suitable silencing target to overcome radioresistance.
Collapse
Affiliation(s)
- Gaia Pucci
- Institute of Molecular Bioimaging and Physiology (IBFM)-National Research Council (CNR), Cefalù Secondary Site, C/da Pietrapollastra-Pisciotto, 90015 Cefalù, Italy; (G.P.); (V.B.); (M.C.); (F.P.C.); (G.R.); (G.I.F.)
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STeBiCeF), University of Palermo, Viale delle Scienze Bld.17, 90128 Palermo, Italy;
| | - Luigi Minafra
- Institute of Molecular Bioimaging and Physiology (IBFM)-National Research Council (CNR), Cefalù Secondary Site, C/da Pietrapollastra-Pisciotto, 90015 Cefalù, Italy; (G.P.); (V.B.); (M.C.); (F.P.C.); (G.R.); (G.I.F.)
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STeBiCeF), University of Palermo, Viale delle Scienze Bld.17, 90128 Palermo, Italy;
| | - Valentina Bravatà
- Institute of Molecular Bioimaging and Physiology (IBFM)-National Research Council (CNR), Cefalù Secondary Site, C/da Pietrapollastra-Pisciotto, 90015 Cefalù, Italy; (G.P.); (V.B.); (M.C.); (F.P.C.); (G.R.); (G.I.F.)
| | - Marco Calvaruso
- Institute of Molecular Bioimaging and Physiology (IBFM)-National Research Council (CNR), Cefalù Secondary Site, C/da Pietrapollastra-Pisciotto, 90015 Cefalù, Italy; (G.P.); (V.B.); (M.C.); (F.P.C.); (G.R.); (G.I.F.)
| | - Giuseppina Turturici
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STeBiCeF), University of Palermo, Viale delle Scienze Bld.17, 90128 Palermo, Italy;
| | - Francesco P. Cammarata
- Institute of Molecular Bioimaging and Physiology (IBFM)-National Research Council (CNR), Cefalù Secondary Site, C/da Pietrapollastra-Pisciotto, 90015 Cefalù, Italy; (G.P.); (V.B.); (M.C.); (F.P.C.); (G.R.); (G.I.F.)
| | - Gaetano Savoca
- Radiation Oncology, ARNAS-Civico Hospital, 90100 Palermo, Italy; (G.S.); (B.A.)
| | - Boris Abbate
- Radiation Oncology, ARNAS-Civico Hospital, 90100 Palermo, Italy; (G.S.); (B.A.)
| | - Giorgio Russo
- Institute of Molecular Bioimaging and Physiology (IBFM)-National Research Council (CNR), Cefalù Secondary Site, C/da Pietrapollastra-Pisciotto, 90015 Cefalù, Italy; (G.P.); (V.B.); (M.C.); (F.P.C.); (G.R.); (G.I.F.)
| | - Vincenzo Cavalieri
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STeBiCeF), University of Palermo, Viale delle Scienze Bld.17, 90128 Palermo, Italy;
| | - Giusi I. Forte
- Institute of Molecular Bioimaging and Physiology (IBFM)-National Research Council (CNR), Cefalù Secondary Site, C/da Pietrapollastra-Pisciotto, 90015 Cefalù, Italy; (G.P.); (V.B.); (M.C.); (F.P.C.); (G.R.); (G.I.F.)
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STeBiCeF), University of Palermo, Viale delle Scienze Bld.17, 90128 Palermo, Italy;
| |
Collapse
|
13
|
Cai Y, Xu H, Xu C, Zuo Z. Adjusting function of camphor on primary metabolism in Cinnamomum camphora stressed by high temperature. PLANT SCIENCE : AN INTERNATIONAL JOURNAL OF EXPERIMENTAL PLANT BIOLOGY 2024; 339:111956. [PMID: 38101618 DOI: 10.1016/j.plantsci.2023.111956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 12/04/2023] [Accepted: 12/09/2023] [Indexed: 12/17/2023]
Abstract
Cinnamomum camphora has great economic value for its wide utilization in traditional medicine and furniture material, and releases lots of monoterpenes to tolerate high temperature. To uncover the adjusting function of monoterpenes on primary metabolism and promoting their utilization as anti-high temperature agents, the photosynthetic capacities, primary metabolite levels, cell ultrastructure and associated gene expression were surveyed in C. camphora when it was blocked monoterpene biosynthesis with fosmidomycin (Fos) and fumigated with camphor (a typical monoterpene in the plant) under high temperature (Fos+38 °C+camphor). Compared with the control (28 °C), high temperature at 38 °C decreased the starch content and starch grain size, and increased the fructose, glucose, sucrose and soluble sugar content. Meanwhile, high temperature also raised the lipid content, with the increase of lipid droplet size and numbers. These variations were further intensified in Fos+ 38 °C treatment. Compared with Fos+ 38 °C treatment, Fos+ 38 °C+camphor treatment improved the starch accumulation by promoting 4 gene expression in starch biosynthesis, and lowered the sugar content by suppressing 3 gene expression in pentose phosphate pathway and promoting 15 gene expression in glycolysis and tricarboxylic acid cycle. Meanwhile, Fos+ 38 °C+camphor treatment also lowered the lipid content, which may be caused by the down-regulation of 2 genes in fatty acid formation and up-regulation of 4 genes in fatty acid decomposition. Although Fos+ 38 °C+camphor treatment improved the photosynthetic capacities in contrast to Fos+ 38 °C treatment, it cannot explain the variations of these primary metabolite levels. Therefore, camphor should adjust related gene expression to maintain the primary metabolism in C. camphora tolerating high temperature.
Collapse
Affiliation(s)
- Yuyan Cai
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou 311300, China; Zhejiang Provincial Key Laboratory of Forest Aromatic Plants-based Healthcare Functions, Zhejiang A&F University, Hangzhou 311300, China; Shanghai Baoshan District Forestry Station, Shanghai 200940, China
| | - Haozhe Xu
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou 311300, China; Zhejiang Provincial Key Laboratory of Forest Aromatic Plants-based Healthcare Functions, Zhejiang A&F University, Hangzhou 311300, China
| | - Chenyi Xu
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou 311300, China; Zhejiang Provincial Key Laboratory of Forest Aromatic Plants-based Healthcare Functions, Zhejiang A&F University, Hangzhou 311300, China
| | - Zhaojiang Zuo
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou 311300, China; Zhejiang Provincial Key Laboratory of Forest Aromatic Plants-based Healthcare Functions, Zhejiang A&F University, Hangzhou 311300, China.
| |
Collapse
|
14
|
Sheikhshabani SH, Modarres P, Ghafouri‐Fard S, Amini‐Farsani Z, Khodaee L, Shaygan N, Amini‐Farsani Z, Omrani MD. Meta-analysis of microarray data to determine gene indicators involved in cisplatin resistance in non-small cell lung cancer. Cancer Rep (Hoboken) 2024; 7:e1970. [PMID: 38351531 PMCID: PMC10864718 DOI: 10.1002/cnr2.1970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 12/02/2023] [Accepted: 12/28/2023] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND Lung cancer is a major cause of cancer-related mortality worldwide, with a 5-year survival rate of approximately 22%. Cisplatin is one of the standard first-line chemotherapeutic agents for non-small cell lung cancer (NSCLC), but its efficacy is often limited by the development of resistance. Despite extensive research on the molecular mechanisms of chemoresistance, the underlying causes remain elusive and complex. AIMS We analyzed three microarray datasets to find the gene signature and key pathways related to cisplatin resistance in NSCLC. METHODS AND RESULTS We compared the gene expression of sensitive and resistant NSCLC cell lines treated with cisplatin. We found 274 DEGs, including 111 upregulated and 163 downregulated genes, in the resistant group. Gene set enrichment analysis showed the potential roles of several DEGs, such as TUBB2B, MAPK7, TUBAL3, MAP2K5, SMUG1, NTHL1, PARP3, NTRK1, G6PD, PDK1, HEY1, YTHDF2, CD274, and MAGEA1, in cisplatin resistance. Functional analysis revealed the involvement of pathways, such as gap junction, base excision repair, central carbon metabolism, and Notch signaling in the resistant cell lines. CONCLUSION We identified several molecular factors that contribute to cisplatin resistance in NSCLC cell lines, involving genes and pathways that regulate gap junction communication, DNA damage repair, ROS balance, EMT induction, and stemness maintenance. These genes and pathways could be targets for future studies to overcome cisplatin resistance in NSCLC.
Collapse
Affiliation(s)
| | - Paratoo Modarres
- Department of Cell and Molecular Biology and Microbiology, Faculty of Science and TechnologyUniversity of IsfahanIsfahanIran
| | - Soudeh Ghafouri‐Fard
- Department of Medical GeneticsShahid Beheshti University of Medical SciencesTehranIran
| | - Zeinab Amini‐Farsani
- Department of Medical GeneticsShahid Beheshti University of Medical SciencesTehranIran
| | - Lavin Khodaee
- Department of Biotechnology and Plant BreedingIslamic Azad University Science and Research BranchTehranIran
| | - Nasibeh Shaygan
- Department of Medical GeneticsShahid Beheshti University of Medical SciencesTehranIran
| | - Zahra Amini‐Farsani
- Bayesian Imaging and Spatial Statistics Group, Institute of StatisticsLudwig‐Maximilian‐Universität MünchenMunichGermany
- Department of StatisticsLorestan UniversityKhorramabadIran
| | - Mir Davood Omrani
- Urogenital Stem Cell Research CenterShahid Beheshti University of Medical SciencesTehranIran
| |
Collapse
|
15
|
Shu ML, Xia JK, Yan J, Feng YJ, Qian CJ, Teng XS, Yao J. Circ_0002395 promotes aerobic glycolysis and proliferation in pancreatic adenocarcinoma cells via miR-548c-3p/PDK1 axis. J Bioenerg Biomembr 2024; 56:55-71. [PMID: 38041751 DOI: 10.1007/s10863-023-09995-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 11/12/2023] [Indexed: 12/03/2023]
Abstract
Circular RNAs (circRNAs) showing unusual expressions have been discovered in pancreatic adenocarcinoma (PAAD). However, the functions and underlying mechanisms of these circRNAs still remain largely unclear. Our current study discovered a notable increase in the expression of circRNA hsa_circ_0002395 (circ_0002395) in both PAAD tissues and cell lines. This up-regulation of circ_0002395 was found to be associated with larger tumor sizes and lymph node metastasis. Furthermore, our findings showed that circ_0002395 facilitated aerobic glycolysis and cell proliferation in PAAD cells by regulating the miR-548c-3p/PDK1 axis. Mechanistically, we identified circ_0002395 as a competing endogenous RNA (ceRNA) that sponged miR-548c-3p, thereby promoting PDK1 expression and aerobic glycolysis, and ultimately resulting in the enhancement of cell proliferation. Our findings found that circ_0002395 promoted proliferation of PAAD cells by enhancing PDK1 expression and aerobic glycolysis by sponging miR-548c-3p.
Collapse
Affiliation(s)
- Meng-Lu Shu
- Early Gastrointestinal Cancer Research Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, 318000, Zhejiang Province, China
- School of Medicine, Taizhou University, Taizhou, 318000, Zhejiang Province, China
| | - Jun-Kai Xia
- Early Gastrointestinal Cancer Research Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, 318000, Zhejiang Province, China
- School of Medicine, Taizhou University, Taizhou, 318000, Zhejiang Province, China
| | - Jing Yan
- Early Gastrointestinal Cancer Research Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, 318000, Zhejiang Province, China
- School of Medicine, Taizhou University, Taizhou, 318000, Zhejiang Province, China
| | - Yu-Jie Feng
- Early Gastrointestinal Cancer Research Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, 318000, Zhejiang Province, China
- School of Medicine, Taizhou University, Taizhou, 318000, Zhejiang Province, China
| | - Cui-Juan Qian
- Early Gastrointestinal Cancer Research Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, 318000, Zhejiang Province, China.
- School of Medicine, Taizhou University, Taizhou, 318000, Zhejiang Province, China.
| | - Xiao-Sheng Teng
- Early Gastrointestinal Cancer Research Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, 318000, Zhejiang Province, China.
- Department of Gastroenterology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, 318000, Zhejiang Province, China.
| | - Jun Yao
- School of Medicine, Taizhou University, Taizhou, 318000, Zhejiang Province, China.
- Department of Gastroenterology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, 318000, Zhejiang Province, China.
| |
Collapse
|
16
|
Zakic T, Kalezic A, Drvendzija Z, Udicki M, Ivkovic Kapicl T, Srdic Galic B, Korac A, Jankovic A, Korac B. Breast Cancer: Mitochondria-Centered Metabolic Alterations in Tumor and Associated Adipose Tissue. Cells 2024; 13:155. [PMID: 38247846 PMCID: PMC10814287 DOI: 10.3390/cells13020155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 01/10/2024] [Accepted: 01/13/2024] [Indexed: 01/23/2024] Open
Abstract
The close cooperation between breast cancer and cancer-associated adipose tissue (CAAT) shapes the malignant phenotype, but the role of mitochondrial metabolic reprogramming and obesity in breast cancer remains undecided, especially in premenopausal women. Here, we examined mitochondrial metabolic dynamics in paired biopsies of malignant versus benign breast tumor tissue and CAAT in normal-weight and overweight/obese premenopausal women. Lower protein level of pyruvate dehydrogenase and citrate synthase in malignant tumor tissue indicated decreased carbon flux from glucose into the Krebs cycle, whereas the trend was just the opposite in malignant CAAT. Simultaneously, stimulated lipolysis in CAAT of obese women was followed by upregulated β-oxidation, as well as fatty acid synthesis enzymes in both tumor tissue and CAAT of women with malignant tumors, corroborating their physical association. Further, protein level of electron transport chain complexes was generally increased in tumor tissue and CAAT from women with malignant tumors, respective to obesity. Preserved mitochondrial structure in malignant tumor tissue was also observed. However, mitochondrial DNA copy number and protein levels of PGC-1α were dependent on both malignancy and obesity in tumor tissue and CAAT. In conclusion, metabolic cooperation between breast cancer and CAAT in premenopausal women involves obesity-related, synchronized changes in mitochondrial metabolism.
Collapse
Affiliation(s)
- Tamara Zakic
- Institute for Biological Research “Sinisa Stankovic”—National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (T.Z.); (A.K.); (A.J.)
| | - Andjelika Kalezic
- Institute for Biological Research “Sinisa Stankovic”—National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (T.Z.); (A.K.); (A.J.)
| | - Zorka Drvendzija
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia; (Z.D.); (M.U.); (B.S.G.)
| | - Mirjana Udicki
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia; (Z.D.); (M.U.); (B.S.G.)
| | - Tatjana Ivkovic Kapicl
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia; (Z.D.); (M.U.); (B.S.G.)
- Oncology Institute of Vojvodina, 21204 Sremska Kamenica, Serbia;
| | - Biljana Srdic Galic
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia; (Z.D.); (M.U.); (B.S.G.)
| | - Aleksandra Korac
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia;
| | - Aleksandra Jankovic
- Institute for Biological Research “Sinisa Stankovic”—National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (T.Z.); (A.K.); (A.J.)
| | - Bato Korac
- Institute for Biological Research “Sinisa Stankovic”—National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (T.Z.); (A.K.); (A.J.)
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia;
| |
Collapse
|
17
|
Nisar H, Labonté FM, Roggan MD, Schmitz C, Chevalier F, Konda B, Diegeler S, Baumstark-Khan C, Hellweg CE. Hypoxia Modulates Radiosensitivity and Response to Different Radiation Qualities in A549 Non-Small Cell Lung Cancer (NSCLC) Cells. Int J Mol Sci 2024; 25:1010. [PMID: 38256084 PMCID: PMC10816011 DOI: 10.3390/ijms25021010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/28/2023] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Hypoxia-induced radioresistance reduces the efficacy of radiotherapy for solid malignancies, including non-small cell lung cancer (NSCLC). Cellular hypoxia can confer radioresistance through cellular and tumor micro-environment adaptations. Until recently, studies evaluating radioresistance secondary to hypoxia were designed to maintain cellular hypoxia only before and during irradiation, while any handling of post-irradiated cells was carried out in standard oxic conditions due to the unavailability of hypoxia workstations. This limited the possibility of simulating in vivo or clinical conditions in vitro. The presence of molecular oxygen is more important for the radiotoxicity of low-linear energy transfer (LET) radiation (e.g., X-rays) than that of high-LET carbon (12C) ions. The mechanisms responsible for 12C ions' potential to overcome hypoxia-induced radioresistance are currently not fully understood. Therefore, the radioresistance of hypoxic A549 NSCLC cells following exposure to X-rays or 12C ions was investigated along with cell cycle progression and gene expression by maintaining hypoxia before, during and after irradiation. A549 cells were incubated under normoxia (20% O2) or hypoxia (1% O2) for 48 h and then irradiated with X-rays (200 kV) or 12C ions (35 MeV/n, LET ~75 keV/µm). Cell survival was evaluated using colony-forming ability (CFA) assays immediately or 24 h after irradiation (late plating). DNA double-strand breaks (DSBs) were analyzed using γH2AX immunofluorescence microscopy. Cell cycle progression was determined by flow cytometry of 4',6-diamidino-2-phenylindole-stained cells. The global transcription profile post-irradiation was evaluated by RNA sequencing. When hypoxia was maintained before, during and after irradiation, hypoxia-induced radioresistance was observed only in late plating CFA experiments. The killing efficiency of 12C ions was much higher than that of X-rays. Cell survival under hypoxia was affected more strongly by the timepoint of plating in the case of X-rays compared to 12C ions. Cell cycle arrest following irradiation under hypoxia was less pronounced but more prolonged. DSB induction and resolution following irradiation were not significantly different under normoxia and hypoxia. Gene expression response to irradiation primarily comprised cell cycle regulation for both radiation qualities and oxygen conditions. Several PI3K target genes involved in cell migration and cell motility were differentially upregulated in hypoxic cells. Hypoxia-induced radioresistance may be linked to altered cell cycle response to irradiation and PI3K-mediated changes in cell motility and migration in A549 cells rather than less DNA damage or faster repair.
Collapse
Affiliation(s)
- Hasan Nisar
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (F.M.L.); (M.D.R.); (C.S.); (B.K.); (S.D.); (C.B.-K.)
- Department of Medical Sciences, Pakistan Institute of Engineering and Applied Sciences (PIEAS), Islamabad 44000, Pakistan
| | - Frederik M. Labonté
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (F.M.L.); (M.D.R.); (C.S.); (B.K.); (S.D.); (C.B.-K.)
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Marie Denise Roggan
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (F.M.L.); (M.D.R.); (C.S.); (B.K.); (S.D.); (C.B.-K.)
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Claudia Schmitz
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (F.M.L.); (M.D.R.); (C.S.); (B.K.); (S.D.); (C.B.-K.)
| | - François Chevalier
- UMR6252 CIMAP, CEA-CNRS-ENSICAEN-University of Caen Normandy, 14000 Caen, France;
| | - Bikash Konda
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (F.M.L.); (M.D.R.); (C.S.); (B.K.); (S.D.); (C.B.-K.)
| | - Sebastian Diegeler
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (F.M.L.); (M.D.R.); (C.S.); (B.K.); (S.D.); (C.B.-K.)
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Christa Baumstark-Khan
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (F.M.L.); (M.D.R.); (C.S.); (B.K.); (S.D.); (C.B.-K.)
| | - Christine E. Hellweg
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (F.M.L.); (M.D.R.); (C.S.); (B.K.); (S.D.); (C.B.-K.)
| |
Collapse
|
18
|
Ahmed SA, Mendonca P, Messeha SS, Oriaku ET, Soliman KFA. The Anticancer Effects of Marine Carotenoid Fucoxanthin through Phosphatidylinositol 3-Kinase (PI3K)-AKT Signaling on Triple-Negative Breast Cancer Cells. Molecules 2023; 29:61. [PMID: 38202644 PMCID: PMC10779870 DOI: 10.3390/molecules29010061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 12/08/2023] [Accepted: 12/12/2023] [Indexed: 01/12/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer that lacks specific targets such as estrogen, progesterone, and HER2 receptors. TNBC affects one in eight women in the United States, making up 15-20% of breast cancer cases. Patients with TNBC can develop resistance to chemotherapy over time, leading to treatment failure. Therefore, finding other options like natural products is necessary for treatment. The advantages of using natural products sourced from plants as anticancer agents are that they are less toxic, more affordable, and have fewer side effects. These products can modulate several cellular processes of the tumor microenvironment, such as proliferation, migration, angiogenesis, cell cycle arrest, and apoptosis. The phosphatidyl inositol 3-kinase (PI3K)-AKT signaling pathway is an important pathway that contributes to the survival and growth of the tumor microenvironment and is associated with these cellular processes. This current study examined the anticancer effects of fucoxanthin, a marine carotenoid isolated from brown seaweed, in the MDA-MB-231 and MDA-MB-468 TNBC cell lines. The methods used in this study include a cytotoxic assay, PI3K-AKT signaling pathway PCR arrays, and Wes analysis. Fucoxanthin (6.25 µM) + TNF-α (50 ng/mL) and TNF-α (50 ng/mL) showed no significant effect on cell viability compared to the control in both MDA-MB-231 and MDA-MB-468 cells after a 24 h treatment period. PI3K-AKT signaling pathway PCR array studies showed that in TNF-α-stimulated (50 ng/mL) MDA-MB-231 and MDA-MB-468 cells, fucoxanthin (6.25 µM) modulated the mRNA expression of 12 genes, including FOXO1, RASA1, HRAS, MAPK3, PDK2, IRS1, EIF4EBP1, EIF4B, PTK2, TIRAP, RHOA, and ELK1. Additionally, fucoxanthin significantly downregulated the protein expression of IRS1, EIF4B, and ELK1 in MDA-MB-231 cells, and no change in the protein expression of EIF4B and ELK1 was shown in MDA-MB-468 cells. Fucoxanthin upregulated the protein expression of RHOA in both cell lines. The modulation of the expression of genes and proteins of the PI3K-AKT signaling pathway may elucidate fucoxanthin's effects in cell cycle progression, apoptotic processes, migration, and proliferation, which shows that PI3K-AKT may be the possible molecular mechanism for fucoxanthin's effects. In conclusion, the results obtained in this study elucidate fucoxanthin's molecular mechanisms and indicate that fucoxanthin may be considered a promising candidate for breast cancer-targeted therapy.
Collapse
Affiliation(s)
- Shade’ A. Ahmed
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA; (S.A.A.); (E.T.O.)
| | - Patricia Mendonca
- Department of Biology, College of Science and Technology, Florida A&M University, Tallahassee, FL 32307, USA;
| | - Samia S. Messeha
- Department of Biology, College of Science and Technology, Florida A&M University, Tallahassee, FL 32307, USA;
| | - Ebenezer T. Oriaku
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA; (S.A.A.); (E.T.O.)
| | - Karam F. A. Soliman
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA; (S.A.A.); (E.T.O.)
| |
Collapse
|
19
|
Li C, Liu C, Zhang J, Lu Y, Jiang B, Xiong H, Li C. Pyruvate dehydrogenase kinase regulates macrophage polarization in metabolic and inflammatory diseases. Front Immunol 2023; 14:1296687. [PMID: 38193078 PMCID: PMC10773690 DOI: 10.3389/fimmu.2023.1296687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 11/13/2023] [Indexed: 01/10/2024] Open
Abstract
Macrophages are highly heterogeneous and plastic, and have two main polarized phenotypes that are determined by their microenvironment, namely pro- and anti-inflammatory macrophages. Activation of pro-inflammatory macrophages is closely associated with metabolic reprogramming, especially that of aerobic glycolysis. Mitochondrial pyruvate dehydrogenase kinase (PDK) negatively regulates pyruvate dehydrogenase complex activity through reversible phosphorylation and further links glycolysis to the tricarboxylic acid cycle and ATP production. PDK is commonly associated with the metabolism and polarization of macrophages in metabolic and inflammatory diseases. This review examines the relationship between PDK and macrophage metabolism and discusses the mechanisms by which PDK regulates macrophage polarization, migration, and inflammatory cytokine secretion in metabolic and inflammatory diseases. Elucidating the relationships between the metabolism and polarization of macrophages under physiological and pathological conditions, as well as the regulatory pathways involved, may provide valuable insights into the etiology and treatment of macrophage-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Chenyu Li
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong, China
| | - Chuanbin Liu
- Department of Pediatric Dentistry, Jining Stomatological Hospital, Jining, Shandong, China
| | - Junfeng Zhang
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong, China
| | - Yanyu Lu
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong, China
| | - Bingtong Jiang
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong, China
| | - Huabao Xiong
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong, China
| | - Chunxia Li
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong, China
| |
Collapse
|
20
|
MaruYama T, Miyazaki H, Komori T, Osana S, Shibata H, Owada Y, Kobayashi S. Curcumin analog GO-Y030 inhibits tumor metastasis and glycolysis. J Biochem 2023; 174:511-518. [PMID: 37656908 PMCID: PMC11002536 DOI: 10.1093/jb/mvad066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/10/2023] [Accepted: 08/24/2023] [Indexed: 09/03/2023] Open
Abstract
Tumor metastasis is one of the worst prognostic features of cancer. Although metastasis is a major cause of cancer-related deaths, an effective treatment has not yet been established. Here, we explore the antitumor effects of GO-Y030, a curcumin analog, via various mechanisms using a mouse model. GO-Y030 treatment of B16-F10 melanoma cells inhibited TGF-β expression and glycolysis. The invasion assay results showed almost complete invasion inhibition following GO-Y030 treatment. Mouse experiments demonstrated that GO-Y030 administration inhibited lung tumor metastasis without affecting vascular endothelial cells. Consistent with this result, GO-Y030 treatment led to the downregulation of MMP2 and VEGFα, inhibiting tumor invasion and metastasis. The silencing of eIF4B, a downstream molecule of S6, attenuated MMP2 expression. Our study demonstrates the novel efficacy of GO-Y030 in inhibiting tumor metastasis by regulating metastasis-associated gene expression via inhibiting dual access, glycolytic and TGF-β pathways.
Collapse
Affiliation(s)
- Takashi MaruYama
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, Seiryo 2-1, Aoba, Sendai, Miyagi, 980-8575, Japan
- Department of Immunology, Akita University, Graduate School of Medicine, Hondo 1-1, Akita, Akita, 010-8543, Japan
| | - Hirofumi Miyazaki
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, Seiryo 2-1, Aoba, Sendai, Miyagi, 980-8575, Japan
| | - Taishi Komori
- Molecular Biology of Bones and Teeth Section, National Institute of Dental and Craniofacial Research(NIDCR), National Institutes of Health, 30 convent drive, Building 30, Bethesda, MD, 20892, USA
| | - Shion Osana
- Department of Engineering Science, University of Electro-Communications, Graduate School of Informatics and Engineering, Chofugaoka 1-5-1, Chofu, Tokyo, 182-8585, Japan
| | - Hiroyuki Shibata
- Department of Clinical Oncology, Akita University, Graduate School of Medicine, Hondo 1-1, Akita, Akita, 010-8543, Japan
| | - Yuji Owada
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, Seiryo 2-1, Aoba, Sendai, Miyagi, 980-8575, Japan
| | - Shuhei Kobayashi
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, Seiryo 2-1, Aoba, Sendai, Miyagi, 980-8575, Japan
| |
Collapse
|
21
|
Farahzadi R, Valipour B, Fathi E, Pirmoradi S, Molavi O, Montazersaheb S, Sanaat Z. Oxidative stress regulation and related metabolic pathways in epithelial-mesenchymal transition of breast cancer stem cells. Stem Cell Res Ther 2023; 14:342. [PMID: 38017510 PMCID: PMC10685711 DOI: 10.1186/s13287-023-03571-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 11/15/2023] [Indexed: 11/30/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a cell remodeling process in which epithelial cells undergo a reversible phenotype switch via the loss of adhesion capacity and acquisition of mesenchymal characteristics. In other words, EMT activation can increase invasiveness and metastatic properties, and prevent the sensitivity of tumor cells to chemotherapeutics, as mesenchymal cells have a higher resistance to chemotherapy and immunotherapy. EMT is orchestrated by a complex and multifactorial network, often linked to episodic, transient, or partial events. A variety of factors have been implicated in EMT development. Based on this concept, multiple metabolic pathways and master transcription factors, such as Snail, Twist, and ZEB, can drive the EMT. Emerging evidence suggests that oxidative stress plays a significant role in EMT induction. One emerging theory is that reducing mitochondrial-derived reactive oxygen species production may contribute to EMT development. This review describes how metabolic pathways and transcription factors are linked to EMT induction and addresses the involvement of signaling pathways.
Collapse
Affiliation(s)
- Raheleh Farahzadi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behnaz Valipour
- Department of Anatomical Sciences, Sarab Faculty of Medical Sciences, Sarab, Iran
| | - Ezzatollah Fathi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Samaneh Pirmoradi
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Ommoleila Molavi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soheila Montazersaheb
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Zohreh Sanaat
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
22
|
Maglov J, Feng MY, Lin D, Barkhouse K, Alexander A, Grbic M, Zhurov V, Grbic V, Tudzarova S. A link between energy metabolism and plant host adaptation states in the two-spotted spider mite, Tetranychus urticae (Koch). Sci Rep 2023; 13:19343. [PMID: 37935795 PMCID: PMC10630510 DOI: 10.1038/s41598-023-46589-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 11/02/2023] [Indexed: 11/09/2023] Open
Abstract
Energy metabolism is a highly conserved process that balances generation of cellular energy and maintenance of redox homeostasis. It consists of five interconnected pathways: glycolysis, tricarboxylic acid cycle, pentose phosphate, trans-sulfuration, and NAD+ biosynthesis pathways. Environmental stress rewires cellular energy metabolism. Type-2 diabetes is a well-studied energy metabolism rewiring state in human pancreatic β-cells where glucose metabolism is uncoupled from insulin secretion. The two-spotted spider mite, Tetranychus urticae (Koch), exhibits a remarkable ability to adapt to environmental stress. Upon transfer to unfavourable plant hosts, mites experience extreme xenobiotic stress that dramatically affects their survivorship and fecundity. However, within 25 generations, mites adapt to the xenobiotic stress and restore their fitness. Mites' ability to withstand long-term xenobiotic stress raises a question of their energy metabolism states during host adaptation. Here, we compared the transcriptional responses of five energy metabolism pathways between host-adapted and non-adapted mites while using responses in human pancreatic islet donors to model these pathways under stress. We found that non-adapted mites and human pancreatic β-cells responded in a similar manner to host plant transfer and diabetogenic stress respectively, where redox homeostasis maintenance was favoured over energy generation. Remarkably, we found that upon host-adaptation, mite energy metabolic states were restored to normal. These findings suggest that genes involved in energy metabolism can serve as molecular markers for mite host-adaptation.
Collapse
Affiliation(s)
- Jorden Maglov
- Department of Biology, The University of Western Ontario, London, N6A 5B7, Canada
| | - Min Yi Feng
- Department of Biology, The University of Western Ontario, London, N6A 5B7, Canada
| | - Dorothy Lin
- Department of Biology, The University of Western Ontario, London, N6A 5B7, Canada
| | - Kennedy Barkhouse
- Department of Biology, The University of Western Ontario, London, N6A 5B7, Canada
| | - Anton Alexander
- Department of Biology, The University of Western Ontario, London, N6A 5B7, Canada
| | - Miodrag Grbic
- Department of Biology, The University of Western Ontario, London, N6A 5B7, Canada
| | - Vladimir Zhurov
- Department of Biology, The University of Western Ontario, London, N6A 5B7, Canada.
| | - Vojislava Grbic
- Department of Biology, The University of Western Ontario, London, N6A 5B7, Canada.
| | - Slavica Tudzarova
- Larry L. Hillblom Islet Research Center, University of California, Los Angeles, CA, 90095, USA.
| |
Collapse
|
23
|
Dong X, Xia S, Du S, Zhu MH, Lai X, Yao SQ, Chen HZ, Fang C. Tumor Metabolism-Rewriting Nanomedicines for Cancer Immunotherapy. ACS CENTRAL SCIENCE 2023; 9:1864-1893. [PMID: 37901179 PMCID: PMC10604035 DOI: 10.1021/acscentsci.3c00702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Indexed: 10/31/2023]
Abstract
Cancer immunotherapy has become an established therapeutic paradigm in oncologic therapy, but its therapeutic efficacy remains unsatisfactory in the majority of cancer patients. Accumulating evidence demonstrates that the metabolically hostile tumor microenvironment (TME), characterized by acidity, deprivation of oxygen and nutrients, and accumulation of immunosuppressive metabolites, promotes the dysfunction of tumor-infiltrating immune cells (TIICs) and thereby compromises the effectiveness of immunotherapy. This indicates the potential role of tumor metabolic intervention in the reinvigoration of antitumor immunity. With the merits of multiple drug codelivery, cell and organelle-specific targeting, controlled drug release, and multimodal therapy, tumor metabolism-rewriting nanomedicines have recently emerged as an attractive strategy to strengthen antitumor immune responses. This review summarizes the current progress in the development of multifunctional tumor metabolism-rewriting nanomedicines for evoking antitumor immunity. A special focus is placed on how these nanomedicines reinvigorate innate or adaptive antitumor immunity by regulating glucose metabolism, amino acid metabolism, lipid metabolism, and nucleotide metabolism at the tumor site. Finally, the prospects and challenges in this emerging field are discussed.
Collapse
Affiliation(s)
- Xiao Dong
- Department
of Pharmacy, School of Medicine, Shanghai
University, Shanghai 200444, China
| | - Shu Xia
- Department
of Pharmacy, School of Medicine, Shanghai
University, Shanghai 200444, China
| | - Shubo Du
- School
of Bioengineering, Dalian University of
Technology, Dalian 116024, China
| | - Mao-Hua Zhu
- Hongqiao
International Institute of Medicine, Tongren Hospital and State Key
Laboratory of Systems Medicine for Cancer, Department of Pharmacology
and Chemical Biology, Shanghai Jiao Tong
University School of Medicine, Shanghai, 200025 China
| | - Xing Lai
- Hongqiao
International Institute of Medicine, Tongren Hospital and State Key
Laboratory of Systems Medicine for Cancer, Department of Pharmacology
and Chemical Biology, Shanghai Jiao Tong
University School of Medicine, Shanghai, 200025 China
| | - Shao Q. Yao
- Department
of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | - Hong-Zhuan Chen
- Institute
of Interdisciplinary Integrative Biomedical Research, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China
| | - Chao Fang
- Hongqiao
International Institute of Medicine, Tongren Hospital and State Key
Laboratory of Systems Medicine for Cancer, Department of Pharmacology
and Chemical Biology, Shanghai Jiao Tong
University School of Medicine, Shanghai, 200025 China
- Key
Laboratory of Basic Pharmacology of Ministry of Education & Joint
International Research Laboratory of Ethnomedicine of Ministry of
Education, Zunyi Medical University, Zunyi 563003, China
| |
Collapse
|
24
|
Wei P, Lin D, Luo C, Zhang M, Deng B, Cui K, Chen Z. High glucose promotes benign prostatic hyperplasia by downregulating PDK4 expression. Sci Rep 2023; 13:17910. [PMID: 37863991 PMCID: PMC10589318 DOI: 10.1038/s41598-023-44954-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 10/13/2023] [Indexed: 10/22/2023] Open
Abstract
As men age, a growing number develop benign prostatic hyperplasia (BPH). According to previous research, diabetes may be a risk factor. Pyruvate dehydrogenase kinase 4 (PDK4) is closely related to glucose metabolism and plays a role in the onset and progression of numerous illnesses. This study aimed to determine the direct effects of high glucose environment on prostate epithelial cells, in particular by altering PDK4 expression levels. In this investigation, normal prostatic epithelial cells (RWPE-1) and human benign prostatic hyperplasia epithelial cells (BPH-1) were treated with 50 mM glucose to show the alteration of high glucose in prostate cells. PDK4-target siRNA, PDK4-expression plasmid were used to investigate the effects of PDK4. Rosiglitazone (RG), a PPARγ agonist, with the potential to up-regulate PDK4 expression was also used for treating prostate cells. The expression of PDK4 in human prostate samples was also analyzed. The effects of high glucose therapy on BPH-1 and RWPE-1 cells were demonstrated to enhance proliferation, epithelial-mesenchymal transition (EMT), suppress apoptosis, and down-regulate PDK4 expression. Additionally, diabetes-related BPH patients had reduced PDK4 expression. Following the application of PDK4-target siRNA, a comparable outcome was seen. The PDK4-expression plasmid therapy, however, produced the opposite results. RG with the ability to elevate PDK4 expression might be used to treat BPH. Changes in the metabolism of lipids and glucose may be the cause of these consequences. These findings showed that high glucose treatment might facilitate BPH development, and may be related to the down-regulation of PDK4. PDK4 might be a potential therapeutic target of BPH.
Collapse
Affiliation(s)
- Pengyu Wei
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Dongxu Lin
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Changcheng Luo
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Mengyang Zhang
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Bolang Deng
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Kai Cui
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Zhong Chen
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
- Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| |
Collapse
|
25
|
Gupta P, Herring B, Kumar N, Telange R, Garcia-Buntley SS, Caceres TW, Colantonio S, Williams F, Kurup P, Carter AM, Lin D, Chen H, Rose B, Jaskula-Sztul R, Mukhtar S, Reddy S, Bibb JA. Faulty Metabolism: A Potential Instigator of an Aggressive Phenotype in Cdk5-dependent Medullary Thyroid Carcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.13.544755. [PMID: 37398342 PMCID: PMC10312670 DOI: 10.1101/2023.06.13.544755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Mechanistic modeling of cancers such as Medullary Thyroid Carcinoma (MTC) to emulate patient-specific phenotypes is challenging. The discovery of potential diagnostic markers and druggable targets in MTC urgently requires clinically relevant animal models. Here we established orthotopic mouse models of MTC driven by aberrantly active Cdk5 using cell-specific promoters. Each of the two models elicits distinct growth differences that recapitulate the less or more aggressive forms of human tumors. The comparative mutational and transcriptomic landscape of tumors revealed significant alterations in mitotic cell cycle processes coupled with the slow-growing tumor phenotype. Conversely, perturbation in metabolic pathways emerged as critical for aggressive tumor growth. Moreover, an overlapping mutational profile was identified between mouse and human tumors. Gene prioritization revealed putative downstream effectors of Cdk5 which may contribute to the slow and aggressive growth in the mouse MTC models. In addition, Cdk5/p25 phosphorylation sites identified as biomarkers for Cdk5-driven neuroendocrine tumors (NETs) were detected in both slow and rapid onset models and were also histologically present in human MTC. Thus, this study directly relates mouse and human MTC models and uncovers vulnerable pathways potentially responsible for differential tumor growth rates. Functional validation of our findings may lead to better prediction of patient-specific personalized combinational therapies.
Collapse
Affiliation(s)
- Priyanka Gupta
- Department of Translational Neuroscience, University of Arizona School of Medicine in Phoenix, Phoenix, AZ 85004-2230, USA
| | - Brendon Herring
- Department of Surgery, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35233, USA
| | - Nilesh Kumar
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Rahul Telange
- Department of Hematology, St Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Sandra S. Garcia-Buntley
- Cancer Research Technology Program, Antibody Characterization Lab, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Tessa W. Caceres
- Cancer Research Technology Program, Antibody Characterization Lab, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Simona Colantonio
- Cancer Research Technology Program, Antibody Characterization Lab, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Ford Williams
- Department of Surgery, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35233, USA
| | - Pradeep Kurup
- Department of Surgery, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35233, USA
| | - Angela M. Carter
- Department of Surgery, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35233, USA
| | - Diana Lin
- Department of Pathology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35233, USA
| | - Herbert Chen
- Department of Surgery, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35233, USA
| | - Bart Rose
- Department of Surgery, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35233, USA
| | - Renata Jaskula-Sztul
- Department of Surgery, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35233, USA
| | - Shahid Mukhtar
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Sushanth Reddy
- Department of Surgery, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35233, USA
| | - James A. Bibb
- Department of Translational Neuroscience, University of Arizona School of Medicine in Phoenix, Phoenix, AZ 85004-2230, USA
| |
Collapse
|
26
|
Nunes-Xavier CE, Emaldi M, Mingo J, Øyjord T, Mælandsmo GM, Fodstad Ø, Errarte P, Larrinaga G, Llarena R, López JI, Pulido R. The expression pattern of pyruvate dehydrogenase kinases predicts prognosis and correlates with immune exhaustion in clear cell renal cell carcinoma. Sci Rep 2023; 13:7339. [PMID: 37147361 PMCID: PMC10162970 DOI: 10.1038/s41598-023-34087-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 04/24/2023] [Indexed: 05/07/2023] Open
Abstract
Renal cancer cells constitute a paradigm of tumor cells with a glycolytic reprogramming which drives metabolic alterations favouring cell survival and transformation. We studied the expression and activity of pyruvate dehydrogenase kinases (PDK1-4), key enzymes of the energy metabolism, in renal cancer cells. We analysed the expression, subcellular distribution and clinicopathological correlations of PDK1-4 by immunohistochemistry of tumor tissue microarray samples from a cohort of 96 clear cell renal cell carcinoma (ccRCC) patients. Gene expression analysis was performed on whole tumor tissue sections of a subset of ccRCC samples. PDK2 and PDK3 protein expression in tumor cells correlated with lower patient overall survival, whereas PDK1 protein expression correlated with higher patient survival. Gene expression analysis revealed molecular association of PDK2 and PDK3 expression with PI3K signalling pathway, as well as with T cell infiltration and exhausted CD8 T cells. Inhibition of PDK by dichloroacetate in human renal cancer cell lines resulted in lower cell viability, which was accompanied by an increase in pAKT. Together, our findings suggest a differential role for PDK enzymes in ccRCC progression, and highlight PDK as actionable metabolic proteins in relation with PI3K signalling and exhausted CD8 T cells in ccRCC.
Collapse
Affiliation(s)
- Caroline E Nunes-Xavier
- Biomarkers in Cancer Unit, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain.
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway.
| | - Maite Emaldi
- Biomarkers in Cancer Unit, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Janire Mingo
- Biomarkers in Cancer Unit, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Tove Øyjord
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Gunhild M Mælandsmo
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
- University of Tromsø - The Arctic University of Norway, Tromsø, Norway
| | - Øystein Fodstad
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Peio Errarte
- Department of Nursing, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Spain
| | - Gorka Larrinaga
- Biomarkers in Cancer Unit, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Department of Nursing, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Spain
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Spain
| | - Roberto Llarena
- Department of Urology, Cruces University Hospital, Barakaldo, Spain
| | - José I López
- Biomarkers in Cancer Unit, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Rafael Pulido
- Biomarkers in Cancer Unit, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain.
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
27
|
Ma H, Sukonina V, Zhang W, Meng F, Subhash S, Palmgren H, Alexandersson I, Han H, Zhou S, Bartesaghi S, Kanduri C, Enerbäck S. The transcription factor Foxp1 regulates aerobic glycolysis in adipocytes and myocytes. J Biol Chem 2023:104795. [PMID: 37150320 DOI: 10.1016/j.jbc.2023.104795] [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: 03/01/2023] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 05/09/2023] Open
Abstract
In recent years, lactate has been recognized as an important circulating energy substrate rather than only a dead-end metabolic waste product generated during glucose oxidation at low levels of oxygen. The term "aerobic glycolysis" has been coined to denote increased glucose uptake and lactate production despite normal oxygen levels and functional mitochondria. Hence, in "aerobic glycolysis" lactate production is a metabolic choice, whereas in "anaerobic glycolysis" it is a metabolic necessity based on inadequate levels of oxygen. Interestingly, lactate can be taken up by cells and oxidized to pyruvate and thus constitutes a source of pyruvate that is independent of insulin. Here, we show that the transcription factor Foxp1 regulates glucose uptake and lactate production in adipocytes and myocytes. Over-expression of Foxp1 leads to increased glucose uptake and lactate production. In addition, protein levels of several enzymes in the glycolytic pathway are upregulated, such as hexokinase 2, phosphofructokinase, aldolase, and lactate dehydrogenase. Using chromatin immunoprecipitation and real-time quantitative PCR (ChIP-qPCR) assays, we demonstrate that Foxp1 directly interacts with promoter consensus cis-elements that regulate expression of several of these target genes. Conversely, knock-down of Foxp1 suppresses these enzyme levels and lowers glucose uptake and lactate production. Moreover, mice with a targeted deletion of Foxp1 in muscle display systemic glucose intolerance with decreased muscle glucose uptake. In primary human adipocytes with induced expression of Foxp1, we find increased glycolysis and glycolytic capacity. Our results indicate Foxp1 may play an important role as a regulator of aerobic glycolysis in adipose tissue and muscle.
Collapse
Affiliation(s)
- Haixia Ma
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, SE405 30 Gothenburg, Sweden
| | - Valentina Sukonina
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, SE405 30 Gothenburg, Sweden
| | - Wei Zhang
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, SE405 30 Gothenburg, Sweden
| | - Fang Meng
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, SE405 30 Gothenburg, Sweden; Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China; Suzhou Institute of Systems Medicine, Suzhou 215123, Jiangsu, China
| | - Santhilal Subhash
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, SE405 30 Gothenburg, Sweden; Karolinska Institutet, Department of Bioscience and Nutrition, Center for Innovative Medicine, Huddinge, Sweden
| | - Henrik Palmgren
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and metabolism (CVRM), BioPharmaceuticals R&D AstraZeneca, Gothenburg, Sweden
| | - Ida Alexandersson
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and metabolism (CVRM), BioPharmaceuticals R&D AstraZeneca, Gothenburg, Sweden
| | - Huiming Han
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, SE405 30 Gothenburg, Sweden; Department of Pathogen Biology, School of Basic Medical Sciences, Beihua University, Jilin, Jilin Province, 132013, China
| | - Shuping Zhou
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, SE405 30 Gothenburg, Sweden; School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Stefano Bartesaghi
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and metabolism (CVRM), BioPharmaceuticals R&D AstraZeneca, Gothenburg, Sweden
| | - Chandrasekhar Kanduri
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, SE405 30 Gothenburg, Sweden
| | - Sven Enerbäck
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, SE405 30 Gothenburg, Sweden.
| |
Collapse
|
28
|
Yang ES, Do Y, Cheon SY, Kim B, Ling J, Cho MK, Kim T, Bae SJ, Ha KT. Andrographolide suppresses aerobic glycolysis and induces apoptotic cell death by inhibiting pyruvate dehydrogenase kinase 1 expression. Oncol Rep 2023; 49:72. [PMID: 36825595 PMCID: PMC9996679 DOI: 10.3892/or.2023.8509] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 02/07/2023] [Indexed: 02/25/2023] Open
Abstract
Metabolic disorder is a major characteristic of cancer cells, and controlling genes involved in metabolic shifts can be an effective strategy for cancer treatment. Andrographolide (AG), a diterpenoid lactone, is widely recognized as a natural anticancer drug due to its ability to inhibit cancer growth. The present study aimed to investigate the mechanism underlying the mitochondrial‑mediated anticancer effect of AG by inhibiting pyruvate dehydrogenase kinase 1 (PDK1) expression in lung cancer cells. Cells were treated with AG and PDK1 mRNA and protein expression was determined using reverse transcription‑quantitative PCR and western blotting, respectively. As a result, AG significantly inhibited the viability of human lung cancer cells and suppressed aerobic glycolysis by decreasing lactate generation. AG further decreased the PDK1 protein and mRNA levels in a dose‑dependent manner. AG‑induced cell death was assessed by flow cytometry and fluorescence microscopy. AG induced apoptotic cell death that was associated with the cleavage of poly (ADP ribose) polymerase, activation of caspase‑3, and mitochondrial damage, which was associated with an increase in reactive oxygen species and loss of mitochondrial membrane potential. AG‑induced cell death was partially suppressed via PDK1 overexpression in lung cancer cells. Therefore, the anticancer effects of AG on human lung cancer cells may negatively regulate the expression of PDK1.
Collapse
Affiliation(s)
- Eun-Sun Yang
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan, Gyeongsangnam 50612, Republic of Korea
| | - Yunju Do
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongsangnam 50612, Republic of Korea
| | - Se-Yun Cheon
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan, Gyeongsangnam 50612, Republic of Korea
| | - Bosung Kim
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongsangnam 50612, Republic of Korea
| | - Jin Ling
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongsangnam 50612, Republic of Korea
| | - Min Kyoung Cho
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan, Gyeongsangnam 50612, Republic of Korea
| | - Taekyung Kim
- Department of Biology Education, Pusan National University, Busan 46241, Republic of Korea
| | - Sung-Jin Bae
- Department of Molecular Biology and Immunology, Kosin University College of Medicine, Busan 49267, Republic of Korea
| | - Ki-Tae Ha
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan, Gyeongsangnam 50612, Republic of Korea
| |
Collapse
|
29
|
Woolbright BL, Rajendran G, Abbott E, Martin A, Didde R, Dennis K, Harris RA, Taylor JA. Pyruvate Dehydrogenase Kinase 4 Deficiency Increases Tumorigenesis in a Murine Model of Bladder Cancer. Cancers (Basel) 2023; 15:1654. [PMID: 36980540 PMCID: PMC10046149 DOI: 10.3390/cancers15061654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/10/2023] Open
Abstract
Pyruvate dehydrogenase kinase 4 (PDK4) is a mitochondrial isozyme in the PDK family (PDK1-4) partially responsible for phosphorylation of pyruvate dehydrogenase (PDH). Phosphorylation of PDH is thought to result in a pro-proliferative shift in metabolism that sustains growth of cancer cells. Previous data from our lab indicate the pan-PDK inhibitor dichloroacetate (DCA) or acute genetic knockdown of PDK4 blocks proliferation of bladder cancer (BCa) cells. The goal of this study was to determine the role of PDK4 in an in vivo BCa model, with the hypothesis that genetic depletion of PDK4 would impair formation of BCa. PDK4-/- or WT animals were exposed to N-Butyl-N-(4-hydroxybutyl) nitrosamine (BBN) for 16 weeks, and tumors were allowed to develop for up to 7 additional weeks. PDK4-/- mice had significantly larger tumors at later time points. When animals were treated with cisplatin, PDK4-/- animals still had larger tumors than WT mice. PDK4 expression was assessed in human tissue and in mice. WT mice lost expression of PDK4 as tumors became muscle-invasive. Similar results were observed in human samples, wherein tumors had less expression of PDK4 than benign tissue. In summary, PDK4 has a complex, multifunctional role in BCa and may represent an underrecognized tumor suppressor.
Collapse
Affiliation(s)
| | - Ganeshkumar Rajendran
- Department of Urology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Erika Abbott
- Department of Urology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Austin Martin
- School of Medicine, Kansas University Medical Center, Kansas City, KS 66160, USA
| | - Ryan Didde
- School of Medicine, Kansas University Medical Center, Kansas City, KS 66160, USA
| | - Katie Dennis
- Department of Pathology, Kansas University Medical Center, Kansas City, KS 66160, USA
| | - Robert A. Harris
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - John A. Taylor
- Department of Urology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
30
|
Xu X, Hou Y, Lin S, Wang K, Ren Y, Zheng T, Zhang X, Li M, Fan L. Sodium selenite inhibits proliferation of lung cancer cells by inhibiting NF-κB nuclear translocation and down-regulating PDK1 expression which is a key enzyme in energy metabolism expression. J Trace Elem Med Biol 2023; 78:127147. [PMID: 36963369 DOI: 10.1016/j.jtemb.2023.127147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 02/21/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023]
Abstract
As a trace element that maintains homeostasis in human body, selenium has significant anti-tumor activity. However, its exact molecular mechanism remains to be elucidated. Sodium selenite (SSe) is the most widely-distributed inorganic selenium in nature. In this study, we selected SSe as the research object to explore its anti-tumor mechanism in lung cancer. In vitro experiment showed that SSe could inhibit the activation of NF-κB signaling pathway, knowing that NF-κB is an important intracellular nuclear transcription factor that regulates the expression of pyruvate dehydrogenase kinase 1 (PDK1), a key energy metabolism switch affecting the survival status of the whole cell.At the same time, Bay11-7082(NF-κB signaling pathway inhibitors) and SSe resulted in phosphorylation of p65 and IκBα, decreased expression of PDK1 and Bcl-2,and increased expression of Bax in lung cancer cells. Our further study demonstrated that the reduction of PDK1 activity inhibited lactate secretion, reduced mitochondrial membrane potential, caused the release of Cytochrome C (Cyto C), activated mitochondrial respiration, and promoted the apoptosis of lung cancer cells. The in vivo experiment revealed that SSe inhibited the activation of NF-κB signaling pathway, decreased the expression of PDK1, and induced lung cancer cell proliferation and apoptosis. All these findings indicated that SSe promoted lung cancer cell apoptosis by inhibiting the activation of NF-κB signaling pathway, down-regulating PDK1 and activating mitochondrial apoptosis pathway.
Collapse
Affiliation(s)
- Xiao Xu
- Institute of Energy Metabolism and Health, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Anhui Provincial Chest Hospital,Hefei 230022, Anhui Province, China
| | - Yaqin Hou
- Institute of Energy Metabolism and Health, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Department of Respiratory Medicine, No. 901 Hospital of the Chinese People's Liberation Army Logistic Support Force, Hefei 230071, Anhui Province, China
| | - Shumeng Lin
- Department of Respiratory Medicine, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Kai Wang
- Department of Respiratory Medicine, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Yanbei Ren
- Department of Respiratory Medicine, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Tiansheng Zheng
- Department of Respiratory Medicine, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Xi Zhang
- The Second People's Hospital of HeFei, Hefei 230011, Anhui Province, China
| | - Ming Li
- Department of Respiratory Medicine, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| | - Lihong Fan
- Institute of Energy Metabolism and Health, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Department of Respiratory Medicine, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| |
Collapse
|
31
|
Chitturi Suryaprakash RT, Shearston K, Farah CS, Fox SA, Iqbal MM, Kadolsky U, Zhong X, Saxena A, Kujan O. A Novel Preclinical In Vitro 3D Model of Oral Carcinogenesis for Biomarker Discovery and Drug Testing. Int J Mol Sci 2023; 24:ijms24044096. [PMID: 36835505 PMCID: PMC9967961 DOI: 10.3390/ijms24044096] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/13/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
This study aimed to develop an in vitro three-dimensional (3D) cell culture model of oral carcinogenesis for the rapid, scalable testing of chemotherapeutic agents. Spheroids of normal (HOK) and dysplastic (DOK) human oral keratinocytes were cultured and treated with 4-nitroquinoline-1-oxide (4NQO). A 3D invasion assay using Matrigel was performed to validate the model. RNA was extracted and subjected to transcriptomic analysis to validate the model and assess carcinogen-induced changes. The VEGF inhibitors pazopanib and lenvatinib were tested in the model and were validated by a 3D invasion assay, which demonstrated that changes induced by the carcinogen in spheroids were consistent with a malignant phenotype. Further validation was obtained by bioinformatic analyses, which showed the enrichment of pathways associated with hallmarks of cancer and VEGF signalling. Overexpression of common genes associated with tobacco-induced oral squamous cell carcinoma (OSCC), such as MMP1, MMP3, MMP9, YAP1, CYP1A1, and CYP1B1, was also observed. Pazopanib and lenvatinib inhibited the invasion of transformed spheroids. In summary, we successfully established a 3D spheroid model of oral carcinogenesis for biomarker discovery and drug testing. This model is a validated preclinical model for OSCC development and would be suitable for testing a range of chemotherapeutic agents.
Collapse
Affiliation(s)
| | - Kate Shearston
- UWA Dental School, The University of Western Australia, Nedlands, WA 6009, Australia
| | - Camile S. Farah
- Australian Centre for Oral Oncology Research and Education, Nedlands, WA 6009, Australia
| | - Simon A. Fox
- UWA Dental School, The University of Western Australia, Nedlands, WA 6009, Australia
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Nedlands, WA 6009, Australia
| | - Muhammad Munir Iqbal
- Genomics WA, Harry Perkins Institute of Medical Research, Telethon Kids Institute, The University of Western Australia, Nedlands, WA 6009, Australia
| | - Ulrich Kadolsky
- Genomics WA, Harry Perkins Institute of Medical Research, Telethon Kids Institute, The University of Western Australia, Nedlands, WA 6009, Australia
| | - Xiao Zhong
- Genomics WA, Harry Perkins Institute of Medical Research, Telethon Kids Institute, The University of Western Australia, Nedlands, WA 6009, Australia
| | - Alka Saxena
- Genomics WA, Harry Perkins Institute of Medical Research, Telethon Kids Institute, The University of Western Australia, Nedlands, WA 6009, Australia
| | - Omar Kujan
- UWA Dental School, The University of Western Australia, Nedlands, WA 6009, Australia
- Correspondence:
| |
Collapse
|
32
|
Thermal Titration Molecular Dynamics (TTMD): Not Your Usual Post-Docking Refinement. Int J Mol Sci 2023; 24:ijms24043596. [PMID: 36835004 PMCID: PMC9968212 DOI: 10.3390/ijms24043596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/01/2023] [Accepted: 02/07/2023] [Indexed: 02/15/2023] Open
Abstract
Molecular docking is one of the most widely used computational approaches in the field of rational drug design, thanks to its favorable balance between the rapidity of execution and the accuracy of provided results. Although very efficient in exploring the conformational degrees of freedom available to the ligand, docking programs can sometimes suffer from inaccurate scoring and ranking of generated poses. To address this issue, several post-docking filters and refinement protocols have been proposed throughout the years, including pharmacophore models and molecular dynamics simulations. In this work, we present the first application of Thermal Titration Molecular Dynamics (TTMD), a recently developed method for the qualitative estimation of protein-ligand unbinding kinetics, to the refinement of docking results. TTMD evaluates the conservation of the native binding mode throughout a series of molecular dynamics simulations performed at progressively increasing temperatures with a scoring function based on protein-ligand interaction fingerprints. The protocol was successfully applied to retrieve the native-like binding pose among a set of decoy poses of drug-like ligands generated on four different pharmaceutically relevant biological targets, including casein kinase 1δ, casein kinase 2, pyruvate dehydrogenase kinase 2, and SARS-CoV-2 main protease.
Collapse
|
33
|
Pavan M, Menin S, Bassani D, Sturlese M, Moro S. Qualitative Estimation of Protein-Ligand Complex Stability through Thermal Titration Molecular Dynamics Simulations. J Chem Inf Model 2022; 62:5715-5728. [PMID: 36315402 PMCID: PMC9709921 DOI: 10.1021/acs.jcim.2c00995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The prediction of ligand efficacy has long been linked to thermodynamic properties such as the equilibrium dissociation constant, which considers both the association and the dissociation rates of a defined protein-ligand complex. In the last 15 years, there has been a paradigm shift, with an increased interest in the determination of kinetic properties such as the drug-target residence time since they better correlate with ligand efficacy compared to other parameters. In this article, we present thermal titration molecular dynamics (TTMD), an alternative computational method that combines a series of molecular dynamics simulations performed at progressively increasing temperatures with a scoring function based on protein-ligand interaction fingerprints for the qualitative estimation of protein-ligand-binding stability. The protocol has been applied to four different pharmaceutically relevant test cases, including protein kinase CK1δ, protein kinase CK2, pyruvate dehydrogenase kinase 2, and SARS-CoV-2 main protease, on a variety of ligands with different sizes, structures, and experimentally determined affinity values. In all four cases, TTMD was successfully able to distinguish between high-affinity compounds (low nanomolar range) and low-affinity ones (micromolar), proving to be a useful screening tool for the prioritization of compounds in a drug discovery campaign.
Collapse
|
34
|
Paramanantham A, Asfiya R, Das S, McCully G, Srivastava A. Extracellular Vesicle (EVs) Associated Non-Coding RNAs in Lung Cancer and Therapeutics. Int J Mol Sci 2022; 23:13637. [PMID: 36362424 PMCID: PMC9655370 DOI: 10.3390/ijms232113637] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 10/26/2022] [Indexed: 08/13/2023] Open
Abstract
Lung cancer is one of the most lethal forms of cancer, with a very high mortality rate. The precise pathophysiology of lung cancer is not well understood, and pertinent information regarding the initiation and progression of lung cancer is currently a crucial area of scientific investigation. Enhanced knowledge about the disease will lead to the development of potent therapeutic interventions. Extracellular vesicles (EVs) are membrane-bound heterogeneous populations of cellular entities that are abundantly produced by all cells in the human body, including the tumor cells. A defined class of EVs called small Extracellular Vesicles (sEVs or exosomes) carries key biomolecules such as RNA, DNA, Proteins and Lipids. Exosomes, therefore, mediate physiological activities and intracellular communication between various cells, including constituent cells of the tumor microenvironment, namely stromal cells, immunological cells, and tumor cells. In recent years, a surge in studying tumor-associated non-coding RNAs (ncRNAs) has been observed. Subsequently, studies have also reported that exosomes abundantly carry different species of ncRNAs and these exosomal ncRNAs are functionally involved in cancer initiation and progression. Here, we discuss the function of exosomal ncRNAs, such as miRNAs and long non-coding RNAs, in the pathophysiology of lung tumors. Further, the future application of exosomal-ncRNAs in clinics as biomarkers and therapeutic targets in lung cancer is also discussed due to the multifaceted influence of exosomes on cellular physiology.
Collapse
Affiliation(s)
- Anjugam Paramanantham
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Rahmat Asfiya
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Siddharth Das
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Grace McCully
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Akhil Srivastava
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO 65212, USA
- Ellis Fischel Cancer Center, University of Missouri School of Medicine, Columbia, MO 65212, USA
| |
Collapse
|
35
|
Choi SYC, Ribeiro CF, Wang Y, Loda M, Plymate SR, Uo T. Druggable Metabolic Vulnerabilities Are Exposed and Masked during Progression to Castration Resistant Prostate Cancer. Biomolecules 2022; 12:1590. [PMID: 36358940 PMCID: PMC9687810 DOI: 10.3390/biom12111590] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 08/27/2023] Open
Abstract
There is an urgent need for exploring new actionable targets other than androgen receptor to improve outcome from lethal castration-resistant prostate cancer. Tumor metabolism has reemerged as a hallmark of cancer that drives and supports oncogenesis. In this regard, it is important to understand the relationship between distinctive metabolic features, androgen receptor signaling, genetic drivers in prostate cancer, and the tumor microenvironment (symbiotic and competitive metabolic interactions) to identify metabolic vulnerabilities. We explore the links between metabolism and gene regulation, and thus the unique metabolic signatures that define the malignant phenotypes at given stages of prostate tumor progression. We also provide an overview of current metabolism-based pharmacological strategies to be developed or repurposed for metabolism-based therapeutics for castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Stephen Y. C. Choi
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Caroline Fidalgo Ribeiro
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, NY 10021, USA
| | - Yuzhuo Wang
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Massimo Loda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, NY 10021, USA
- New York Genome Center, New York, NY 10013, USA
| | - Stephen R. Plymate
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, 850 Republican St., Seattle, WA 98109, USA
- Geriatrics Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Takuma Uo
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, 850 Republican St., Seattle, WA 98109, USA
| |
Collapse
|
36
|
Genus Smenospongia: Untapped Treasure of Biometabolites—Biosynthesis, Synthesis, and Bioactivities. Molecules 2022; 27:molecules27185969. [PMID: 36144705 PMCID: PMC9501515 DOI: 10.3390/molecules27185969] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/10/2022] [Accepted: 09/10/2022] [Indexed: 11/16/2022] Open
Abstract
Marine sponges continue to attract remarkable attention as one of the richest pools of bioactive metabolites in the marine environment. The genus Smenospongia (order Dictyoceratida, family Thorectidae) sponges can produce diverse classes of metabolites with unique and unusual chemical skeletons, including terpenoids (sesqui-, di-, and sesterterpenoids), indole alkaloids, aplysinopsins, bisspiroimidazolidinones, chromenes, γ-pyrones, phenyl alkenes, naphthoquinones, and polyketides that possessed diversified bioactivities. This review provided an overview of the reported metabolites from Smenospongia sponges, including their biosynthesis, synthesis, and bioactivities in the period from 1980 to June 2022. The structural characteristics and diverse bioactivities of these metabolites could attract a great deal of attention from natural-product chemists and pharmaceuticals seeking to develop these metabolites into medicine for the treatment and prevention of certain health concerns.
Collapse
|
37
|
Ito N, Takatsu A, Ito H, Koike Y, Yoshioka K, Kamei Y, Imai SI. Slc12a8 in the lateral hypothalamus maintains energy metabolism and skeletal muscle functions during aging. Cell Rep 2022; 40:111131. [PMID: 35905718 DOI: 10.1016/j.celrep.2022.111131] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 05/01/2022] [Accepted: 07/01/2022] [Indexed: 12/20/2022] Open
Abstract
Sarcopenia and frailty are urgent socio-economic problems worldwide. Here we demonstrate a functional connection between the lateral hypothalamus (LH) and skeletal muscle through Slc12a8, a recently identified nicotinamide mononucleotide transporter, and its relationship to sarcopenia and frailty. Slc12a8-expressing cells are mainly localized in the LH. LH-specific knockdown of Slc12a8 in young mice decreases activity-dependent energy and carbohydrate expenditure and skeletal muscle functions, including muscle mass, muscle force, intramuscular glycolysis, and protein synthesis. LH-specific Slc12a8 knockdown also decreases sympathetic nerve signals at neuromuscular junctions and β2-adrenergic receptors in skeletal muscle, indicating the importance of the LH-sympathetic nerve-β2-adrenergic receptor axis. LH-specific overexpression of Slc12a8 in aged mice significantly ameliorates age-associated decreases in energy expenditure and skeletal muscle functions. Our results highlight an important role of Slc12a8 in the LH for regulation of whole-body metabolism and skeletal muscle functions and provide insights into the pathogenesis of sarcopenia and frailty during aging.
Collapse
Affiliation(s)
- Naoki Ito
- AMED Frailty Research Laboratory (Teijin), AMED Cyclic Innovation for Clinical Empowerment (CiCLE), Osaka, Japan; Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation (IBRI), Foundation for Biomedical Research and Innovation (FBRI), Kobe, Japan
| | - Ai Takatsu
- AMED Frailty Research Laboratory (Teijin), AMED Cyclic Innovation for Clinical Empowerment (CiCLE), Osaka, Japan; Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation (IBRI), Foundation for Biomedical Research and Innovation (FBRI), Kobe, Japan
| | - Hiromi Ito
- AMED Frailty Research Laboratory (Teijin), AMED Cyclic Innovation for Clinical Empowerment (CiCLE), Osaka, Japan; Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation (IBRI), Foundation for Biomedical Research and Innovation (FBRI), Kobe, Japan
| | - Yuka Koike
- AMED Frailty Research Laboratory (Teijin), AMED Cyclic Innovation for Clinical Empowerment (CiCLE), Osaka, Japan; Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation (IBRI), Foundation for Biomedical Research and Innovation (FBRI), Kobe, Japan
| | - Kiyoshi Yoshioka
- Institute for Research on Productive Aging (IRPA), Tokyo, Japan; Laboratory of Molecular Nutrition, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan
| | - Yasutomi Kamei
- Laboratory of Molecular Nutrition, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan
| | - Shin-Ichiro Imai
- AMED Frailty Research Laboratory (Teijin), AMED Cyclic Innovation for Clinical Empowerment (CiCLE), Osaka, Japan; Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation (IBRI), Foundation for Biomedical Research and Innovation (FBRI), Kobe, Japan; Department of Developmental Biology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
38
|
Yao X, Zhang Q. Function and Clinical Significance of Circular RNAs in Thyroid Cancer. Front Mol Biosci 2022; 9:925389. [PMID: 35936780 PMCID: PMC9353217 DOI: 10.3389/fmolb.2022.925389] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/22/2022] [Indexed: 12/28/2022] Open
Abstract
Thyroid cancer (TC) is the leading cause and mortality of endocrine malignancies worldwide. Tumourigenesis involves multiple molecules including circular RNAs (circRNAs). circRNAs with covalently closed single-stranded structures have been identified as a type of regulatory RNA because of their high stability, abundance, and tissue/developmental stage-specific expression. Accumulating evidence has demonstrated that various circRNAs are aberrantly expressed in thyroid tissues, cells, exosomes, and body fluids in patients with TC. CircRNAs have been identified as either oncogenic or tumour suppressor roles in regulating tumourigenesis, tumour metabolism, metastasis, ferroptosis, and chemoradiation resistance in TC. Importantly, circRNAs exert pivotal effects on TC through various mechanisms, including acting as miRNA sponges or decoys, interacting with RNA-binding proteins, and translating functional peptides. Recent studies have suggested that many different circRNAs are associated with certain clinicopathological features, implying that the altered expression of circRNAs may be characteristic of TC. The purpose of this review is to provide an overview of recent advances on the dysregulation, functions, molecular mechanisms and potential clinical applications of circRNAs in TC. This review also aimes to improve our understanding of the functions of circRNAs in the initiation and progression of cancer, and to discuss the future perspectives on strategies targeting circRNAs in TC.
Collapse
|
39
|
Nunes-Xavier CE, Mingo J, Emaldi M, Flem-Karlsen K, Mælandsmo GM, Fodstad Ø, Llarena R, López JI, Pulido R. Heterogeneous Expression and Subcellular Localization of Pyruvate Dehydrogenase Complex in Prostate Cancer. Front Oncol 2022; 12:873516. [PMID: 35692804 PMCID: PMC9174590 DOI: 10.3389/fonc.2022.873516] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/31/2022] [Indexed: 12/02/2022] Open
Abstract
Background Pyruvate dehydrogenase (PDH) complex converts pyruvate into acetyl-CoA by pyruvate decarboxylation, which drives energy metabolism during cell growth, including prostate cancer (PCa) cell growth. The major catalytic subunit of PDH, PDHA1, is regulated by phosphorylation/dephosphorylation by pyruvate dehydrogenase kinases (PDKs) and pyruvate dehydrogenase phosphatases (PDPs). There are four kinases, PDK1, PDK2, PDK3 and PDK4, which can phosphorylate and inactivate PDH; and two phosphatases, PDP1 and PDP2, that dephosphorylate and activate PDH. Methods We have analyzed by immunohistochemistry the expression and clinicopathological correlations of PDHA1, PDP1, PDP2, PDK1, PDK2, PDK3, and PDK4, as well as of androgen receptor (AR), in a retrospective PCa cohort of patients. A total of 120 PCa samples of representative tumor areas from all patients were included in tissue microarray (TMA) blocks for analysis. In addition, we studied the subcellular localization of PDK2 and PDK3, and the effects of the PDK inhibitor dichloroacetate (DCA) in the growth, proliferation, and mitochondrial respiration of PCa cells. Results We found heterogeneous expression of the PDH complex components in PCa tumors. PDHA1, PDP1, PDK1, PDK2, and PDK4 expression correlated positively with AR expression. A significant correlation of PDK2 immunostaining with biochemical recurrence and disease-free survival was revealed. In PCa tissue specimens, PDK2 displayed cytoplasmic and nuclear immunostaining, whereas PDK1, PDK3 and PDK4 showed mostly cytoplasmic staining. In cells, ectopically expressed PDK2 and PDK3 were mainly localized in mitochondria compartments. An increase in maximal mitochondrial respiration was observed in PCa cells upon PDK inhibition by DCA, in parallel with less proliferative capacity. Conclusion Our findings support the notion that expression of specific PDH complex components is related with AR signaling in PCa tumors. Furthermore, PDK2 expression associated with poor PCa prognosis. This highlights a potential for PDH complex components as targets for intervention in PCa.
Collapse
Affiliation(s)
- Caroline E Nunes-Xavier
- Biomarkers in Cancer, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain.,Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Janire Mingo
- Biomarkers in Cancer, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Maite Emaldi
- Biomarkers in Cancer, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Karine Flem-Karlsen
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Gunhild M Mælandsmo
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Øystein Fodstad
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Roberto Llarena
- Department of Urology, Cruces University Hospital, Barakaldo, Spain
| | - José I López
- Biomarkers in Cancer, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain.,Department of Pathology, Cruces University Hospital, Barakaldo, Spain
| | - Rafael Pulido
- Biomarkers in Cancer, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain.,Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
40
|
Khodaei T, Inamdar S, Suresh AP, Acharya AP. Drug delivery for metabolism targeted cancer immunotherapy. Adv Drug Deliv Rev 2022; 184:114242. [PMID: 35367306 DOI: 10.1016/j.addr.2022.114242] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/26/2022] [Accepted: 03/26/2022] [Indexed: 02/08/2023]
Abstract
Drug delivery vehicles have made a great impact on cancer immunotherapies in clinics and pre-clinical research. Notably, the science of delivery of cancer vaccines and immunotherapeutics, modulating immune cell functions has inspired development of several successful companies and clinical products. Interestingly, these drug delivery modalities not only modulate the function of immune cells (often quantified at the mRNA and protein levels), but also modulate the metabolism of these cells. Specifically, cancer immunotherapy often leads to activation of different immune cells such as dendritic cells, macrophages and T cells, which is driven by energy metabolism of these cells. Recently, there has been a great excitement about interventions that can directly modulate the energy metabolism of these immune cells and thus affect their function and in turn lead to a robust cancer immune response. Here we review few strategies that have been tested in clinic and pre-clinical research for generating effective metabolism-associated cancer therapies and immunotherapies.
Collapse
|
41
|
Holmes AG, Parker JB, Sagar V, Truica MI, Soni PN, Han H, Schiltz GE, Abdulkadir SA, Chakravarti D. A MYC inhibitor selectively alters the MYC and MAX cistromes and modulates the epigenomic landscape to regulate target gene expression. SCIENCE ADVANCES 2022; 8:eabh3635. [PMID: 35476451 PMCID: PMC9045724 DOI: 10.1126/sciadv.abh3635] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 03/11/2022] [Indexed: 06/14/2023]
Abstract
MYC regulates multiple gene programs, raising questions about the potential selectivity and downstream transcriptional consequences of MYC inhibitors as cancer therapeutics. Here, we examined the effect of a small-molecule MYC inhibitor, MYCi975, on the MYC/MAX cistromes, epigenome, transcriptome, and tumorigenesis. Integrating these data revealed three major classes of MYCi975-modulated gene targets: type 1 (down-regulated), type 2 (up-regulated), and type 3 (unaltered). While cell cycle and signal transduction pathways were heavily targeted by MYCi, RNA biogenesis and core transcriptional pathway genes were spared. MYCi975 altered chromatin binding of MYC and the MYC network family proteins, and chromatin accessibility and H3K27 acetylation alterations revealed MYCi975 suppression of MYC-regulated lineage factors AR/ARv7, FOXA1, and FOXM1. Consequently, MYCi975 synergistically sensitized resistant prostate cancer cells to enzalutamide and estrogen receptor-positive breast cancer cells to 4-hydroxytamoxifen. Our results demonstrate that MYCi975 selectively inhibits MYC target gene expression and provide a mechanistic rationale for potential combination therapies.
Collapse
Affiliation(s)
- Austin G. Holmes
- Division of Reproductive Sciences in Medicine, Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - J. Brandon Parker
- Division of Reproductive Sciences in Medicine, Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Vinay Sagar
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Mihai I. Truica
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Pritin N. Soni
- Division of Reproductive Sciences in Medicine, Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Huiying Han
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Gary E. Schiltz
- Department of Chemistry, Northwestern University, Evanston, IL 60208, USA
- The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Sarki A. Abdulkadir
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Debabrata Chakravarti
- Division of Reproductive Sciences in Medicine, Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
42
|
PDK4 Constitutes a Novel Prognostic Biomarker and Therapeutic Target in Gastric Cancer. Diagnostics (Basel) 2022; 12:diagnostics12051101. [PMID: 35626257 PMCID: PMC9139696 DOI: 10.3390/diagnostics12051101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/05/2022] [Accepted: 04/20/2022] [Indexed: 02/04/2023] Open
Abstract
Gastric cancer (GC) is one of the most prevalent and deadly malignancies worldwide. We aimed to assess the functional role and clinical significance of pyruvate dehydrogenase kinase (PDK) in GC and explored the underlying mechanisms. The bioinformatics method was used to investigate the expression of PDKs in GC, the effect on clinical outcomes, enriched pathways, interactive network, and the correlation between PDK4 and immune infiltration. Next, PDK expression in the GC cells and tissues were verified by qRT-PCR and western blotting. A Cell Counting Kit-8 (CCK8), colony-formation, Flow cytometry, Transwell and wound healing assays were carried out to evaluate the influence of PDK4 on cell proliferation, invasion and migration. Among PDKs, PDK4 expression was aberrant in GC and identified as an independent prognostic factor. GO analysis, GSEA, and PPI showed that PDK4 expression may regulate cell adhesion, metal ion transport, synaptic activity, and cancer cell metabolism in GC. Analyses of immune infiltration showed that PDK4 correlated with the abundant expression of various immunocytes. Finally, we verified that upregulation of PDK4 expression enhanced the ability of GC cells to proliferate, migrate, and invade. In conclusion, PDK4 was identified as a potential candidate diagnostic biomarker and therapeutic target for GC patients.
Collapse
|
43
|
miR-32 promotes MYC-driven prostate cancer. Oncogenesis 2022; 11:11. [PMID: 35228520 PMCID: PMC8885642 DOI: 10.1038/s41389-022-00385-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 12/24/2022] Open
Abstract
miR-32 is an androgen receptor (AR)-regulated microRNA, expression of which is increased in castration-resistant prostate cancer (PC). We have previously shown that overexpression of miR-32 in the prostate of transgenic mice potentiates proliferation in prostate epithelium. Here, we set out to determine whether increased expression of miR-32 influences growth or phenotype in prostate adenocarcinoma in vivo. We studied transgenic mice expressing MYC oncogene (hiMYC mice) to induce tumorigenesis in the mouse prostate and discovered that transgenic overexpression of miR-32 resulted in increased tumor burden as well as a more aggressive tumor phenotype in this model. Elevated expression of miR-32 increased proliferation as assessed by Ki-67 immunohistochemistry, increased nuclear density, and higher mitotic index in the tumors. By gene expression analysis of the tumorous prostate tissue, we confirmed earlier findings that miR-32 expression regulates prostate secretome by modulating expression levels of several PC-related target genes such as Spink1, Spink5, and Msmb. Further, we identified Pdk4 as a tumor-associated miR-32 target in the mouse prostate. Expression analysis of PDK4 in human PC reveals an inverse correlation with miR-32 expression and Gleason score, a decrease in castration-resistant and metastatic tumors compared to untreated primary PC, and an association of low PDK4 expression with a shorter recurrence-free survival of patients. Although decreased PDK4 expression induces the higher metabolic activity of PC cells, induced expression of PDK4 reduces both mitotic respiration and glycolysis rates as well as inhibits cell growth. In conclusion, we show that miR-32 promotes MYC-induced prostate adenocarcinoma and identifies PDK4 as a PC-relevant metabolic target of miR-32-3p.
Collapse
|
44
|
Park JH, Lee HK. Current Understanding of Hypoxia in Glioblastoma Multiforme and Its Response to Immunotherapy. Cancers (Basel) 2022; 14:1176. [PMID: 35267480 PMCID: PMC8909860 DOI: 10.3390/cancers14051176] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 02/20/2022] [Accepted: 02/23/2022] [Indexed: 02/06/2023] Open
Abstract
Hypoxia is a hallmark of glioblastoma multiforme (GBM), the most aggressive cancer of the central nervous system, and is associated with multiple aspects of tumor pathogenesis. For example, hypoxia induces resistance to conventional cancer therapies and inhibits antitumor immune responses. Thus, targeting hypoxia is an attractive strategy for GBM therapy. However, traditional studies on hypoxia have largely excluded the immune system. Recently, the critical role of the immune system in the defense against multiple tumors has become apparent, leading to the development of effective immunotherapies targeting numerous cancer types. Critically, however, GBM is classified as a "cold tumor" due to poor immune responses. Thus, to improve GBM responsiveness against immunotherapies, an improved understanding of both immune function in GBM and the role of hypoxia in mediating immune responses within the GBM microenvironment is needed. In this review, we discuss the role of hypoxia in GBM from a clinical, pathological, and immunological perspective.
Collapse
Affiliation(s)
| | - Heung Kyu Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea;
| |
Collapse
|
45
|
Cevatemre B, Ulukaya E, Dere E, Dilege S, Acilan C. Pyruvate Dehydrogenase Contributes to Drug Resistance of Lung Cancer Cells Through Epithelial Mesenchymal Transition. Front Cell Dev Biol 2022; 9:738916. [PMID: 35083212 PMCID: PMC8785343 DOI: 10.3389/fcell.2021.738916] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 12/13/2021] [Indexed: 01/10/2023] Open
Abstract
Recently, there has been a growing interest on the role of mitochondria in metastatic cascade. Several reports have shown the preferential utilization of glycolytic pathway instead of mitochondrial respiration for energy production and the pyruvate dehydrogenase (PDH) has been considered to be a contributor to this switch in some cancers. Since epithelial mesenchymal transition (EMT) is proposed to be one of the significant mediators of metastasis, the molecular connections between cancer cell metabolism and EMT may reveal underlying mechanisms and improve our understanding on metastasis. In order to explore a potential role for PDH inhibition on EMT and associated drug resistance, we took both pharmacological and genetic approaches, and selectively inhibited or knocked down PDHA1 by using Cpi613 and shPDHA1, respectively. We found that both approaches triggered morphological changes and characteristics of EMT (increase in mesenchymal markers). This change was accompanied by enhanced wound healing and an increase in migration. Interestingly, cells were more resistant to many of the clinically used chemotherapeutics following PDH inhibition or PDHA1 knockdown. Furthermore, the TGFβRI (known as a major inducer of the EMT) inhibitor (SB-431542) together with the PDHi, was effective in reversing EMT. In conclusion, interfering with PDH induced EMT, and more importantly resulted in chemoresistance. Therefore, our study demonstrates the need for careful consideration of PDH-targeting approaches in cancer treatment.
Collapse
Affiliation(s)
- Buse Cevatemre
- Research Center for Translational Medicine, Koc University, Istanbul, Turkey.,Department of Biology, Uludag University, Bursa, Turkey
| | - Engin Ulukaya
- Department of Clinical Biochemistry, Istinye University Faculty of Medicine, Istanbul, Turkey
| | - Egemen Dere
- Department of Biology, Uludag University, Bursa, Turkey
| | - Sukru Dilege
- School of Medicine, Koc University, Istanbul, Turkey
| | - Ceyda Acilan
- Research Center for Translational Medicine, Koc University, Istanbul, Turkey.,School of Medicine, Koc University, Istanbul, Turkey
| |
Collapse
|
46
|
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
|
47
|
Zhang Y, Zhu B, Cai Y, Zhu S, Zhao H, Ying X, Jiang C, Zeng J. Alteration in glycolytic/cholesterogenic gene expression is associated with bladder cancer prognosis and immune cell infiltration. BMC Cancer 2022; 22:2. [PMID: 34980012 PMCID: PMC8722165 DOI: 10.1186/s12885-021-09064-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 11/26/2021] [Indexed: 12/12/2022] Open
Abstract
Background Oncogenic metabolic reprogramming contributes to tumor growth and immune evasion. The intertumoral metabolic heterogeneity and interaction of distinct metabolic pathways may determine patient outcomes. In this study, we aim to determine the clinical and immunological significance of metabolic subtypes according to the expression levels of genes related to glycolysis and cholesterol-synthesis in bladder cancer (BCa). Methods Based on the median expression levels of glycolytic and cholesterogenic genes, patients were stratified into 4 subtypes (mixed, cholesterogenic, glycolytic, and quiescent) in an integrated cohort including TCGA, GSE13507, and IMvigor210. Clinical, genomic, transcriptomic, and tumor microenvironment characteristics were compared between the 4 subtypes. Results The 4 metabolic subtypes exhibited distinct clinical, molecular, and genomic patterns. Compared to quiescent subtype, mixed subtype was more likely to be basal tumors and was significantly associated with poorer prognosis even after controlling for age, gender, histological grade, clinical stage, and molecular phenotypes. Additionally, mixed tumors harbored a higher frequency of RB1 and LRP1B copy number deletion compared to quiescent tumors (25.7% vs. 12.7 and 27.9% vs. 10.2%, respectively, both adjusted P value< 0.05). Furthermore, aberrant PIK3CA expression level was significantly correlated with those of glycolytic and cholesterogenic genes. The quiescent subtype was associated with lower stemness indices and lower signature scores for gene sets involved in genomic instability, including DNA replication, DNA damage repair, mismatch repair, and homologous recombination genes. Moreover, quiescent tumors exhibited lower expression levels of pyruvate dehydrogenase kinases 1-3 (PDK1-3) than the other subtypes. In addition, distinct immune cell infiltration patterns were observed across the 4 metabolic subtypes, with greater infiltration of M0/M2 macrophages observed in glycolytic and mixed subtypes. However, no significant difference in immunotherapy response was observed across the 4 metabolic subtypes. Conclusion This study proposed a new metabolic subtyping method for BCa based on genes involved in glycolysis and cholesterol synthesis pathways. Our findings may provide novel insight for the development of personalized subtype-specific treatment strategies targeting metabolic vulnerabilities. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-09064-0.
Collapse
Affiliation(s)
- Yuying Zhang
- Department of Urology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China.,Department of Obstetrics, Shenzhen Longhua Maternity and Child Healthcare Hospital, Shenzhen, 510089, China
| | - Baoyi Zhu
- Department of Urology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Yi Cai
- Department of Urology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha, 410008, China
| | - Sihua Zhu
- Department of Urology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Hongjun Zhao
- Department of Urology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Xiaoling Ying
- Department of Translational Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 51000, China
| | - Chonghe Jiang
- Department of Urology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Jianwen Zeng
- Department of Urology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China.
| |
Collapse
|
48
|
Chen CL, Lin CY, Kung HJ. Targeting Mitochondrial OXPHOS and Their Regulatory Signals in Prostate Cancers. Int J Mol Sci 2021; 22:13435. [PMID: 34948229 PMCID: PMC8708687 DOI: 10.3390/ijms222413435] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/13/2021] [Accepted: 12/13/2021] [Indexed: 12/26/2022] Open
Abstract
Increasing evidence suggests that tumor development requires not only oncogene/tumor suppressor mutations to drive the growth, survival, and metastasis but also metabolic adaptations to meet the increasing energy demand for rapid cellular expansion and to cope with the often nutritional and oxygen-deprived microenvironment. One well-recognized strategy is to shift the metabolic flow from oxidative phosphorylation (OXPHOS) or respiration in mitochondria to glycolysis or fermentation in cytosol, known as Warburg effects. However, not all cancer cells follow this paradigm. In the development of prostate cancer, OXPHOS actually increases as compared to normal prostate tissue. This is because normal prostate epithelial cells divert citrate in mitochondria for the TCA cycle to the cytosol for secretion into seminal fluid. The sustained level of OXPHOS in primary tumors persists in progression to an advanced stage. As such, targeting OXPHOS and mitochondrial activities in general present therapeutic opportunities. In this review, we summarize the recent findings of the key regulators of the OXPHOS pathway in prostate cancer, ranging from transcriptional regulation, metabolic regulation to genetic regulation. Moreover, we provided a comprehensive update of the current status of OXPHOS inhibitors for prostate cancer therapy. A challenge of developing OXPHOS inhibitors is to selectively target cancer mitochondria and spare normal counterparts, which is also discussed.
Collapse
Affiliation(s)
- Chia-Lin Chen
- Ph.D. Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; (C.-L.C.); (C.-Y.L.)
| | - Ching-Yu Lin
- Ph.D. Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; (C.-L.C.); (C.-Y.L.)
| | - Hsing-Jien Kung
- Ph.D. Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; (C.-L.C.); (C.-Y.L.)
- Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 110, Taiwan
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli County 350, Taiwan
- Comprehensive Cancer Center, Department of Biochemistry and Molecular Medicine, University of California at Davis, Sacramento, CA 95817, USA
| |
Collapse
|
49
|
Zheng X, Xu H, Yi X, Zhang T, Wei Q, Li H, Ai J. Tumor-antigens and immune landscapes identification for prostate adenocarcinoma mRNA vaccine. Mol Cancer 2021; 20:160. [PMID: 34872584 PMCID: PMC8645679 DOI: 10.1186/s12943-021-01452-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 10/27/2021] [Indexed: 02/08/2023] Open
Abstract
Prostate adenocarcinoma (PRAD) is a leading cause of death among men. Messenger ribonucleic acid (mRNA) vaccine presents an attractive approach to achieve satisfactory outcomes; however, tumor antigen screening and vaccination candidates show a bottleneck in this field. We aimed to investigate the tumor antigens for mRNA vaccine development and immune subtypes for choosing appropriate patients for vaccination. We identified eight overexpressed and mutated tumor antigens with poor prognostic value of PRAD, including KLHL17, CPT1B, IQGAP3, LIME1, YJEFN3, KIAA1529, MSH5 and CELSR3. The correlation of those genes with antigen-presenting immune cells were assessed. We further identified three immune subtypes of PRAD (PRAD immune subtype [PIS] 1-3) with distinct clinical, molecular, and cellular characteristics. PIS1 showed better survival and immune cell infiltration, nevertheless, PIS2 and PIS3 showed cold tumor features with poorer prognosis and higher tumor genomic instability. Moreover, these immune subtypes presented distinguished association with immune checkpoints, immunogenic cell death modulators, and prognostic factors of PRAD. Furthermore, immune landscape characterization unraveled the immune heterogeneity among patients with PRAD. To summarize, our study suggests KLHL17, CPT1B, IQGAP3, LIME1, YJEFN3, KIAA1529, MSH5 and CELSR3 are potential antigens for PRAD mRNA vaccine development, and patients in the PIS2 and PIS3 groups are more suitable for vaccination.
Collapse
Affiliation(s)
- Xiaonan Zheng
- Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Institute of Systems Genetics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hang Xu
- Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xianyanling Yi
- Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Tianyi Zhang
- Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qiang Wei
- Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hong Li
- Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jianzhong Ai
- Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Department of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
50
|
Wiebringhaus R, Pecoraro M, Neubauer HA, Trachtová K, Trimmel B, Wieselberg M, Pencik J, Egger G, Krall C, Moriggl R, Mann M, Hantusch B, Kenner L. Proteomic Analysis Identifies NDUFS1 and ATP5O as Novel Markers for Survival Outcome in Prostate Cancer. Cancers (Basel) 2021; 13:6036. [PMID: 34885151 PMCID: PMC8656993 DOI: 10.3390/cancers13236036] [Citation(s) in RCA: 1] [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: 10/12/2021] [Revised: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 12/12/2022] Open
Abstract
We aimed to identify novel markers for aggressive prostate cancer in a STAT3-low proteomics-derived dataset of mitochondrial proteins by immunohistochemical analysis and correlation with transcriptomic data and biochemical recurrence in a STAT3 independent PCa cohort. Formalin-fixed paraffin-embedded tissue (FFPE) sample selection for proteomic analysis and tissue-microarray (TMA) generation was conducted from a cohort of PCa patients. Retrospective data analysis was performed with the same cohort. 153 proteins differentially expressed between STAT3-low and STAT3-high samples were identified. Out of these, 46 proteins were associated with mitochondrial processes including oxidative phosphorylation (OXPHOS), and 45 proteins were upregulated, including NDUFS1/ATP5O. In a STAT3 independent PCa cohort, high expression of NDUFS1/ATP5O was confirmed by immunocytochemistry (IHC) and was significantly associated with earlier biochemical recurrence (BCR). mRNA expression levels for these two genes were significantly higher in intra-epithelial neoplasia and in PCa compared to benign prostate glands. NDUFS1/ATP5O levels are increased both at the mRNA and protein level in aggressive PCa. Our results provide evidence that NDUFS1/ATP5O could be used to identify high-risk PCa patients.
Collapse
Affiliation(s)
- Robert Wiebringhaus
- Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria; (B.T.); (M.W.); (J.P.); (G.E.); (B.H.)
- Department of Otolaryngology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Matteo Pecoraro
- Institute for Research in Biomedicine, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland;
| | - Heidi A. Neubauer
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (H.A.N.); (R.M.)
| | - Karolína Trachtová
- Central European Institute of Technology, Masaryk University, 60177 Brno, Czech Republic;
- Christian Doppler Laboratory for Applied Metabolomics (CDL-AM), Medical University of Vienna, 1090 Vienna, Austria
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Bettina Trimmel
- Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria; (B.T.); (M.W.); (J.P.); (G.E.); (B.H.)
| | - Maritta Wieselberg
- Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria; (B.T.); (M.W.); (J.P.); (G.E.); (B.H.)
| | - Jan Pencik
- Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria; (B.T.); (M.W.); (J.P.); (G.E.); (B.H.)
| | - Gerda Egger
- Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria; (B.T.); (M.W.); (J.P.); (G.E.); (B.H.)
- Ludwig Boltzmann Institute Applied Diagnostics, 1090 Vienna, Austria
| | - Christoph Krall
- Institute for Statistics, Medical University of Vienna, 1090 Vienna, Austria;
| | - Richard Moriggl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (H.A.N.); (R.M.)
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany;
| | - Brigitte Hantusch
- Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria; (B.T.); (M.W.); (J.P.); (G.E.); (B.H.)
| | - Lukas Kenner
- Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria; (B.T.); (M.W.); (J.P.); (G.E.); (B.H.)
- Christian Doppler Laboratory for Applied Metabolomics (CDL-AM), Medical University of Vienna, 1090 Vienna, Austria
- Center for Biomarker Research in Medicine (CBmed), 8010 Graz, Austria
- Unit for Laboratory Animal Pathology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| |
Collapse
|