51
|
Chen X, Abdallah MF, Grootaert C, Van Nieuwerburgh F, Rajkovic A. New insights into the combined toxicity of aflatoxin B1 and fumonisin B1 in HepG2 cells using Seahorse respirometry analysis and RNA transcriptome sequencing. ENVIRONMENT INTERNATIONAL 2023; 175:107945. [PMID: 37126917 DOI: 10.1016/j.envint.2023.107945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 04/08/2023] [Accepted: 04/19/2023] [Indexed: 05/03/2023]
Abstract
Aflatoxin B1 (AFB1) and fumonisin B1 (FB1) are widely (co-)detected in food and known for their hepatotoxicity in humans. Still, their combined toxicity needs to be investigated, especially the impact on mitochondria. In our previous work, we examined the effect of short-term exposure to different doses of AFB1, FB1, and their binary mixture (MIX) on the bioenergetic status of HepG2 cells, a well-recognized in vitro model system for studying liver cell function. In the current work, we further investigated the (combined) effect of AFB1 and FB1 on the mitochondrial and glycolytic activity of HepG2 cells using Seahorse respirometry analysis and RNA transcriptome sequencing. The results showed that the co-exposure, especially at high doses, is more toxic due to a more inhibition of all parameters of mitochondrial respiration. However, FB1 contributes more to the MIX effects than AFB1. RNA transcriptome sequencing showed that the p53 signaling pathway, a major orchestrator of mitochondrial apoptosis, was differentially expressed. Moreover, the co-exposure significantly downregulated the genes encoding for Complexes I, II, III, and IV, representing the onset of the suppressed mitochondrial respiration in HepG2 cells.
Collapse
Affiliation(s)
- Xiangrong Chen
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium.
| | - Mohamed F Abdallah
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium; Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| | - Charlotte Grootaert
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Filip Van Nieuwerburgh
- Laboratory of Pharmaceutical Biotechnology, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Andreja Rajkovic
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium.
| |
Collapse
|
52
|
Niu N, Ye J, Hu Z, Zhang J, Wang Y. Regulative Roles of Metabolic Plasticity Caused by Mitochondrial Oxidative Phosphorylation and Glycolysis on the Initiation and Progression of Tumorigenesis. Int J Mol Sci 2023; 24:ijms24087076. [PMID: 37108242 PMCID: PMC10139088 DOI: 10.3390/ijms24087076] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/23/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
One important feature of tumour development is the regulatory role of metabolic plasticity in maintaining the balance of mitochondrial oxidative phosphorylation and glycolysis in cancer cells. In recent years, the transition and/or function of metabolic phenotypes between mitochondrial oxidative phosphorylation and glycolysis in tumour cells have been extensively studied. In this review, we aimed to elucidate the characteristics of metabolic plasticity (emphasizing their effects, such as immune escape, angiogenesis migration, invasiveness, heterogeneity, adhesion, and phenotypic properties of cancers, among others) on tumour progression, including the initiation and progression phases. Thus, this article provides an overall understanding of the influence of abnormal metabolic remodeling on malignant proliferation and pathophysiological changes in carcinoma.
Collapse
Affiliation(s)
- Nan Niu
- Shenzhen Engineering Labortaory for Marine Algal Biotechnology, Longhua Innovation Institute for Biotechnology, College of Life Sciences and Oceanography, Lihu Campus of Shenzhen University, Shenzhen 518055, China
- College of Physics and Optoelectronic Engineering, Canghai Campus of Shenzhen University, Shenzhen 518060, China
| | - Jinfeng Ye
- Shenzhen Engineering Labortaory for Marine Algal Biotechnology, Longhua Innovation Institute for Biotechnology, College of Life Sciences and Oceanography, Lihu Campus of Shenzhen University, Shenzhen 518055, China
| | - Zhangli Hu
- Shenzhen Engineering Labortaory for Marine Algal Biotechnology, Longhua Innovation Institute for Biotechnology, College of Life Sciences and Oceanography, Lihu Campus of Shenzhen University, Shenzhen 518055, China
| | - Junbin Zhang
- Shenzhen Engineering Labortaory for Marine Algal Biotechnology, Longhua Innovation Institute for Biotechnology, College of Life Sciences and Oceanography, Lihu Campus of Shenzhen University, Shenzhen 518055, China
| | - Yun Wang
- Shenzhen Engineering Labortaory for Marine Algal Biotechnology, Longhua Innovation Institute for Biotechnology, College of Life Sciences and Oceanography, Lihu Campus of Shenzhen University, Shenzhen 518055, China
| |
Collapse
|
53
|
Marcucci F, Rumio C. On the Role of Glycolysis in Early Tumorigenesis-Permissive and Executioner Effects. Cells 2023; 12:cells12081124. [PMID: 37190033 DOI: 10.3390/cells12081124] [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: 02/01/2023] [Revised: 03/26/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
Reprogramming energy production from mitochondrial respiration to glycolysis is now considered a hallmark of cancer. When tumors grow beyond a certain size they give rise to changes in their microenvironment (e.g., hypoxia, mechanical stress) that are conducive to the upregulation of glycolysis. Over the years, however, it has become clear that glycolysis can also associate with the earliest steps of tumorigenesis. Thus, many of the oncoproteins most commonly involved in tumor initiation and progression upregulate glycolysis. Moreover, in recent years, considerable evidence has been reported suggesting that upregulated glycolysis itself, through its enzymes and/or metabolites, may play a causative role in tumorigenesis, either by acting itself as an oncogenic stimulus or by facilitating the appearance of oncogenic mutations. In fact, several changes induced by upregulated glycolysis have been shown to be involved in tumor initiation and early tumorigenesis: glycolysis-induced chromatin remodeling, inhibition of premature senescence and induction of proliferation, effects on DNA repair, O-linked N-acetylglucosamine modification of target proteins, antiapoptotic effects, induction of epithelial-mesenchymal transition or autophagy, and induction of angiogenesis. In this article we summarize the evidence that upregulated glycolysis is involved in tumor initiation and, in the following, we propose a mechanistic model aimed at explaining how upregulated glycolysis may play such a role.
Collapse
Affiliation(s)
- Fabrizio Marcucci
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Trentacoste 2, 20134 Milan, Italy
| | - Cristiano Rumio
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Trentacoste 2, 20134 Milan, Italy
| |
Collapse
|
54
|
A reduced model of cell metabolism to revisit the glycolysis-OXPHOS relationship in the deregulated tumor microenvironment. J Theor Biol 2023; 562:111434. [PMID: 36739944 DOI: 10.1016/j.jtbi.2023.111434] [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: 07/19/2022] [Revised: 01/24/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
Cancer cells metabolism focuses the interest of the cancer research community. Although this process is intensely studied experimentally, there are very few theoretical models that address this issue. One of the main reasons is the extraordinary complexity of the metabolism that involves numerous interdependent regulatory networks which makes the computational recreation of this complexity illusory. In this study we propose a reduced model of the metabolism which focuses on the interrelation of the three main energy metabolites which are oxygen, glucose and lactate in order to better understand the dynamics of the core system of the glycolysis-OXPHOS relationship. So simple as it is, the model highlights the main rules allowing the cell to dynamically adapt its metabolism to its changing environment. It also makes it possible to address this impact at the tissue scale. The simulations carried out in a spheroid show non-trivial spatial heterogeneity of energy metabolism. It further suggests that the metabolic features that are commonly attributed to cancer cells are not necessarily due to an intrinsic abnormality of the cells. They can emerge spontaneously due to the deregulated over-acidic environment.
Collapse
|
55
|
Fennell EMJ, Aponte-Collazo LJ, Pathmasiri W, Rushing BR, Barker NK, Partridge MC, Li YY, White CA, Greer YE, Herring LE, Lipkowitz S, Sumner SCJ, Iwanowicz EJ, Graves LM. Multi-omics analyses reveal ClpP activators disrupt essential mitochondrial pathways in triple-negative breast cancer. Front Pharmacol 2023; 14:1136317. [PMID: 37063293 PMCID: PMC10103842 DOI: 10.3389/fphar.2023.1136317] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 02/27/2023] [Indexed: 04/03/2023] Open
Abstract
ClpP activators ONC201 and related small molecules (TR compounds, Madera Therapeutics), have demonstrated significant anti-cancer potential in vitro and in vivo studies, including clinical trials for refractory solid tumors. Though progress has been made in identifying specific phenotypic outcomes following ClpP activation, the exact mechanism by which ClpP activation leads to broad anti-cancer activity has yet to be fully elucidated. In this study, we utilized a multi-omics approach to identify the ClpP-dependent proteomic, transcriptomic, and metabolomic changes resulting from ONC201 or the TR compound TR-57 in triple-negative breast cancer cells. Applying mass spectrometry-based methods of proteomics and metabolomics, we identified ∼8,000 proteins and 588 metabolites, respectively. From proteomics data, 113 (ONC201) and 191 (TR-57) proteins significantly increased and 572 (ONC201) and 686 (TR-57) proteins significantly decreased in this study. Gene ontological (GO) analysis revealed strong similarities between proteins up- or downregulated by ONC201 or TR-57 treatment. Notably, this included the downregulation of many mitochondrial processes and proteins, including mitochondrial translation and mitochondrial matrix proteins. We performed a large-scale transcriptomic analysis of WT SUM159 cells, identifying ∼7,700 transcripts (746 and 1,100 significantly increasing, 795 and 1,013 significantly decreasing in ONC201 and TR-57 treated cells, respectively). Less than 21% of these genes were affected by these compounds in ClpP null cells. GO analysis of these data demonstrated additional similarity of response to ONC201 and TR-57, including a decrease in transcripts related to the mitochondrial inner membrane and matrix, cell cycle, and nucleus, and increases in other nuclear transcripts and transcripts related to metal-ion binding. Comparison of response between both compounds demonstrated a highly similar response in all -omics datasets. Analysis of metabolites also revealed significant similarities between ONC201 and TR-57 with increases in α-ketoglutarate and 2-hydroxyglutaric acid and decreased ureidosuccinic acid, L-ascorbic acid, L-serine, and cytidine observed following ClpP activation in TNBC cells. Further analysis identified multiple pathways that were specifically impacted by ClpP activation, including ATF4 activation, heme biosynthesis, and the citrulline/urea cycle. In summary the results of our studies demonstrate that ONC201 and TR-57 induce highly similar and broad effects against multiple mitochondrial processes required for cell proliferation.
Collapse
Affiliation(s)
- Emily M. J. Fennell
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Lucas J. Aponte-Collazo
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Wimal Pathmasiri
- Department of Nutrition, Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, United States
| | - Blake R. Rushing
- Department of Nutrition, Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, United States
| | - Natalie K. Barker
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Michael Hooker Proteomics Core Facility, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Megan C. Partridge
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Yuan-Yuan Li
- Department of Nutrition, Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, United States
| | - Cody A. White
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Yoshimi E. Greer
- Women’s Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Laura E. Herring
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Michael Hooker Proteomics Core Facility, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Stanley Lipkowitz
- Women’s Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Susan C. J. Sumner
- Department of Nutrition, Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, United States
| | | | - Lee M. Graves
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- *Correspondence: Lee M. Graves,
| |
Collapse
|
56
|
Moreno-Sánchez R, Robledo-Cadena DX, Pacheco-Velázquez SC, Vargas Navarro JL, Padilla-Flores JA, Rodríguez-Enríquez S. Estimation of energy pathway fluxes in cancer cells - Beyond the Warburg effect. Arch Biochem Biophys 2023; 739:109559. [PMID: 36906097 DOI: 10.1016/j.abb.2023.109559] [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: 10/24/2022] [Revised: 02/15/2023] [Accepted: 03/04/2023] [Indexed: 03/11/2023]
Abstract
Glycolytic and respiratory fluxes were analyzed in cancer and non-cancer cells. The steady-state fluxes in energy metabolism were used to estimate the contributions of aerobic glycolytic and oxidative phosphorylation (OxPhos) pathways to the cellular ATP supply. The rate of lactate production - corrected for the fraction generated by glutaminolysis - is proposed as the appropriate way to estimate glycolytic flux. In general, the glycolytic rates estimated for cancer cells are higher than those found in non-cancer cells, as originally observed by Otto Warburg. The rate of basal or endogenous cellular O2 consumption corrected for non-ATP synthesizing O2 consumption, measured after inhibition by oligomycin (a specific, potent and permeable ATP synthase inhibitor), has been proposed as the appropriate way to estimate mitochondrial ATP synthesis-linked O2 flux or net OxPhos flux in living cells. Detecting non-negligible oligomycin-sensitive O2 consumption rates in cancer cells has revealed that the mitochondrial function is not impaired, as claimed by the Warburg effect. Furthermore, when calculating the relative contributions to cellular ATP supply, under a variety of environmental conditions and for different types of cancer cells, it was found that OxPhos pathway was the main ATP provider over glycolysis. Hence, OxPhos pathway targeting can be successfully used to block in cancer cells ATP-dependent processes such as migration. These observations may guide the re-design of novel targeted therapies.
Collapse
Affiliation(s)
- Rafael Moreno-Sánchez
- Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Carrera de Biología, Laboratorio de Control Metabólico, Los Reyes Ixtacala, Hab. Los Reyes Ixtacala Barrio de los Árboles/Barrio de los Héroes, Tlalnepantla, 54090, Mexico.
| | | | | | - Jorge Luis Vargas Navarro
- Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Carrera de Biología, Laboratorio de Control Metabólico, Los Reyes Ixtacala, Hab. Los Reyes Ixtacala Barrio de los Árboles/Barrio de los Héroes, Tlalnepantla, 54090, Mexico
| | - Joaquín Alberto Padilla-Flores
- Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Carrera de Biología, Laboratorio de Control Metabólico, Los Reyes Ixtacala, Hab. Los Reyes Ixtacala Barrio de los Árboles/Barrio de los Héroes, Tlalnepantla, 54090, Mexico
| | - Sara Rodríguez-Enríquez
- Instituto Nacional de Cardiología, Departamento de Bioquímica, Ciudad de México, 14080, Mexico; Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Carrera de Medicina, Laboratorio de Control Metabólico, Los Reyes Ixtacala, Hab. Los Reyes Ixtacala Barrio de los Árboles/Barrio de los Héroes, Tlalnepantla, 54090, Mexico.
| |
Collapse
|
57
|
Li SP, Ou L, Zhang Y, Shen FR, Chen YG. A first-in-class POLRMT specific inhibitor IMT1 suppresses endometrial carcinoma cell growth. Cell Death Dis 2023; 14:152. [PMID: 36823110 PMCID: PMC9950144 DOI: 10.1038/s41419-023-05682-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/25/2023]
Abstract
Exploring novel molecularly-targeted therapies for endometrial carcinoma is important. The current study explored the potential anti-endometrial carcinoma activity by a first-in-class POLRMT (RNA polymerase mitochondrial) inhibitor IMT1. In patient-derived primary human endometrial carcinoma cells and established lines, treatment with IMT1 potently inhibited cell viability, proliferation, cell-cycle progression and motility, while inducing robust caspase-apoptosis activation. Treatment with the PLORMT inhibitor impaired mitochondrial functions, leading to mtDNA (mitochondrial DNA) transcription inhibition, mitochondrial membrane potential decline, reactive oxygen species formation, oxidative stress and ATP loss in the endometrial carcinoma cells. Similarly, POLRMT depletion, through shRNA-induced silencing or CRISPR/Cas9-caused knockout (KO), inhibited primary endometrial carcinoma cell proliferation and motility, and induced mitochondrial dysfunction and apoptosis. Importantly, IMT1 failed to induce further cytotoxicity in POLRMT-KO endometrial carcinoma cells. Contrarily, ectopic overexpression of POLRMT further augmented proliferation and motility of primary endometrial carcinoma cells. In vivo, oral administration of a single dose of IMT1 substantially inhibited endometrial carcinoma xenograft growth in the nude mice. mtDNA transcription inhibition, oxidative stress, ATP loss and apoptosis were detected in IMT1-treated endometrial carcinoma xenograft tissues. Together, targeting PLORMT by IMT1 inhibited endometrial carcinoma cell growth in vitro and in vivo.
Collapse
Affiliation(s)
- Shu-ping Li
- grid.429222.d0000 0004 1798 0228Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, China ,grid.89957.3a0000 0000 9255 8984Obstetrics Department, The Affiliated Changzhou No.2 People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Li Ou
- grid.452666.50000 0004 1762 8363Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yan Zhang
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China.
| | - Fang-rong Shen
- grid.429222.d0000 0004 1798 0228Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - You-guo Chen
- grid.429222.d0000 0004 1798 0228Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
58
|
Zheng Y, Zhang J, Zhu X, Wei Y, Zhao W, Si S, Li Y. A Mitochondrial Perspective on Noncommunicable Diseases. Biomedicines 2023; 11:biomedicines11030647. [PMID: 36979626 PMCID: PMC10045938 DOI: 10.3390/biomedicines11030647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/05/2023] [Accepted: 02/16/2023] [Indexed: 02/24/2023] Open
Abstract
Mitochondria are the center of energy metabolism in eukaryotic cells and play a central role in the metabolism of living organisms. Mitochondrial diseases characterized by defects in oxidative phosphorylation are the most common congenital diseases. Meanwhile, mitochondrial dysfunction caused by secondary factors such as non-inherited genetic mutations can affect normal physiological functions of human cells, induce apoptosis, and lead to the development of various diseases. This paper reviewed several major factors and mechanisms that contribute to mitochondrial dysfunction and discussed the development of diseases closely related to mitochondrial dysfunction and drug treatment strategies discovered in recent years.
Collapse
Affiliation(s)
- Yifan Zheng
- Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jing Zhang
- Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xiaohong Zhu
- Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yuanjuan Wei
- Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Wuli Zhao
- NHC Key Laboratory of Antibiotic Bioengineering, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Correspondence: (W.Z.); (S.S.); (Y.L.)
| | - Shuyi Si
- Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Correspondence: (W.Z.); (S.S.); (Y.L.)
| | - Yan Li
- Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Correspondence: (W.Z.); (S.S.); (Y.L.)
| |
Collapse
|
59
|
Zhang Y, Zhou F, Guan J, Zhou L, Chen B. Action Mechanism of Metformin and Its Application in Hematological Malignancy Treatments: A Review. Biomolecules 2023; 13:250. [PMID: 36830619 PMCID: PMC9953052 DOI: 10.3390/biom13020250] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 02/03/2023] Open
Abstract
Hematologic malignancies (HMs) mainly include acute and chronic leukemia, lymphoma, myeloma and other heterogeneous tumors that seriously threaten human life and health. The common effective treatments are radiotherapy, chemotherapy and hematopoietic stem cell transplantation (HSCT), which have limited options and are prone to tumor recurrence and (or) drug resistance. Metformin is the first-line drug for the treatment of type 2 diabetes (T2DM). Recently, studies identified the potential anti-cancer ability of metformin in both T2DM patients and patients that are non-diabetic. The latest epidemiological and preclinical studies suggested a potential benefit of metformin in the prevention and treatment of patients with HM. The mechanism may involve the activation of the adenosine monophosphate-activated protein kinase (AMPK) signaling pathway by metformin as well as other AMPK-independent pathways to exert anti-cancer properties. In addition, combining current conventional anti-cancer drugs with metformin may improve the efficacy and reduce adverse drug reactions. Therefore, metformin can also be used as an adjuvant therapeutic agent for HM. This paper highlights the anti-hyperglycemic effects and potential anti-cancer effects of metformin, and also compiles the in vitro and clinical trials of metformin as an anti-cancer and chemosensitizing agent for the treatment of HM. The need for future research on the use of metformin in the treatment of HM is indicated.
Collapse
Affiliation(s)
| | | | | | | | - Baoan Chen
- Department of Hematology and Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| |
Collapse
|
60
|
Rybkowska P, Radoszkiewicz K, Kawalec M, Dymkowska D, Zabłocka B, Zabłocki K, Sarnowska A. The Metabolic Changes between Monolayer (2D) and Three-Dimensional (3D) Culture Conditions in Human Mesenchymal Stem/Stromal Cells Derived from Adipose Tissue. Cells 2023; 12:cells12010178. [PMID: 36611971 PMCID: PMC9818744 DOI: 10.3390/cells12010178] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/22/2022] [Accepted: 12/30/2022] [Indexed: 01/03/2023] Open
Abstract
INTRODUCTION One of the key factors that may influence the therapeutic potential of mesenchymal stem/stromal cells (MSCs) is their metabolism. The switch between mitochondrial respiration and glycolysis can be affected by many factors, including the oxygen concentration and the spatial form of culture. This study compared the metabolic features of adipose-derived mesenchymal stem/stromal cells (ASCs) and dedifferentiated fat cells (DFATs) cultivated as monolayer or spheroid culture under 5% O2 concentration (physiological normoxia) and their impact on MSCs therapeutic abilities. RESULTS We observed that the cells cultured as spheroids had a slightly lower viability and a reduced proliferation rate but a higher expression of the stemness-related transcriptional factors compared to the cells cultured in monolayer. The three-dimensional culture form increased mtDNA content, oxygen consumption rate (OCR) and extracellular acidification rate (ECAR), especially in DFATs-3D population. The DFATs spheroids also demonstrated increased levels of Complex V proteins and higher rates of ATP production. Moreover, increased reactive oxygen species and lower intracellular lactic acid levels were also found in 3D culture. CONCLUSION Our results may suggest that metabolic reconfiguration accompanies the transition from 2D to 3D culture and the processes of both mitochondrial respiration and glycolysis become more active. Intensified metabolism might be associated with the increased demand for energy, which is needed to maintain the expression of pluripotency genes and stemness state.
Collapse
Affiliation(s)
- Paulina Rybkowska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Klaudia Radoszkiewicz
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Maria Kawalec
- Molecular Biology Unit, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Dorota Dymkowska
- Laboratory of Cellular Metabolism, Nencki Institute of Experimental Biology, Polish Academy of Science, 02-093 Warsaw, Poland
| | - Barbara Zabłocka
- Molecular Biology Unit, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Krzysztof Zabłocki
- Laboratory of Cellular Metabolism, Nencki Institute of Experimental Biology, Polish Academy of Science, 02-093 Warsaw, Poland
| | - Anna Sarnowska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
- Correspondence: ; Tel.: +48-22-608-6598
| |
Collapse
|
61
|
Alteration of Cellular Energy Metabolism through LPAR2-Axin2 Axis in Gastric Cancer. Biomolecules 2022; 12:biom12121805. [PMID: 36551233 PMCID: PMC9775664 DOI: 10.3390/biom12121805] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/29/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
Lysophosphatidic acid (LPA), a multifunctional endogenous phospholipid, plays a vital role in cellular homeostasis and the malignant behavior of cancer cells through G-protein-coupled receptors. However, the role of LPA in β-catenin-mediated gastric cancer is unknown. Here, we have noted the high expression of LPAR2 in human gastric cancer tissues, and that LPA treatment significantly increased the proliferation, migration, and invasion of human gastric cancer cells. Results from our biochemical experiments showed that an LPA exposure increased the expression of β-catenin and its nuclear localization, increased the phosphorylation of glycogen synthase kinase 3β (GSK-3β), decreased the expression of Axin2, and increased the expression of the target genes of the β-catenin signaling pathway. The LPA2 receptor (LPAR2) antagonist significantly reduced the LPA-induced nuclear localization of β-catenin, the primary signaling event. The knockdown of LPAR2 in the gastric cancer cell lines robustly reduced the LPA-induced β-catenin activity. An LPA exposure increased the ATP production by both oxidative phosphorylation and glycolysis, and this effect was abrogated with the addition of an LPAR2 antagonist and XAV393, which stabilizes the Axin and inhibits the β-catenin signaling pathway. Based on our findings, the possibility that LPA contributes to gastric cancer initiation and progression through the β-catenin signaling pathway as well as by the dysregulation of the energy metabolism via the LPAR2 receptor and Axin2, respectively, provides a novel insight into the mechanism of and possible therapeutic targets of gastric cancer.
Collapse
|
62
|
Zhang M, Du M, Qi X, Wang Y, Li G, Xu C, Zhang X. Retro-inversion follicle-stimulating hormone peptide-modified nanoparticles for delivery of PDK2 shRNA against chemoresistant ovarian cancer by switching glycolysis to oxidative phosphorylation. Cancer Nanotechnol 2022. [DOI: 10.1186/s12645-022-00129-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Most ovarian cancers are diagnosed at advanced stages characterized by abdominal dissemination and frequently exhibit chemoresistance. Pyruvate dehydrogenase kinase 2 (PDK2) regulates the switch between glycolysis and oxidative phosphorylation and contributes to tumor progression and chemoresistance. Here, we investigated the effects of PDK2 blockade on metabolic reprogramming and cisplatin sensitivity and evaluated the in vivo antitumor effects of PDK2 shRNA in chemoresistant ovarian cancer using retro-inverso follicle-stimulating hormone peptide-modified nanoparticle as carriers.
Methods
The expression of PDK2 was detected by immunohistochemistry, Western blot and real-time PCR. Cell proliferation and apoptosis were detected using CCK-8 and flow cytometry. Cell migration was detected by Transwell assay. Seahorse Analyzer was used to evaluate metabolic changes. The cisplatin-resistant ovarian cancer cells A2780cp were used to establish the mouse model of peritoneal metastatic ovarian cancer.
Results
A higher expression level of PDK2 was observed in chemoresistant ovarian cancer tissues and cell lines and was associated with shorter progression-free survival. PDK2 knockdown inhibited proliferation and migration and promoted apoptosis of both cisplatin-sensitive and cisplatin-resistant ovarian cancer cells. Cisplatin sensitivity was increased even in cisplatin-resistant ovarian cancer cells. Mechanistically, PDK2 knockdown resulted in an increased oxygen consumption rate and decreased extracellular acidification rate, along with reduced lactate production, increased PDHC activity and increased levels of electron transport chain complexes III and V. The metabolism switched from glycolysis to oxidative phosphorylation. Finally, to specifically and effectively deliver PDK2 shRNA in vivo, we formulated a targeted delivery system containing retro-inverso follicle-stimulating hormone peptide as a targeting moiety and polyethylene glycol–polyethylenimine copolymers as carriers. The nanoparticle complex significantly suppressed tumor growth and peritoneal metastasis of cisplatin-resistant ovarian cancer without obvious toxicities.
Conclusions
Our findings showed the link between metabolic reprogramming and chemoresistance in ovarian cancer and provided an effective targeting strategy for switching metabolic pathways in cancer therapy.
Collapse
|
63
|
An Update on the Metabolic Landscape of Oncogenic Viruses. Cancers (Basel) 2022; 14:cancers14235742. [PMID: 36497226 PMCID: PMC9738352 DOI: 10.3390/cancers14235742] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/10/2022] [Accepted: 11/17/2022] [Indexed: 11/24/2022] Open
Abstract
Viruses play an important role in cancer development as about 12% of cancer types are linked to viral infections. Viruses that induce cellular transformation are known as oncoviruses. Although the mechanisms of viral oncogenesis differ between viruses, all oncogenic viruses share the ability to establish persistent chronic infections with no obvious symptoms for years. During these prolonged infections, oncogenic viruses manipulate cell signaling pathways that control cell cycle progression, apoptosis, inflammation, and metabolism. Importantly, it seems that most oncoviruses depend on these changes for their persistence and amplification. Metabolic changes induced by oncoviruses share many common features with cancer metabolism. Indeed, viruses, like proliferating cancer cells, require increased biosynthetic precursors for virion production, need to balance cellular redox homeostasis, and need to ensure host cell survival in a given tissue microenvironment. Thus, like for cancer cells, viral replication and persistence of infected cells frequently depend on metabolic changes. Here, we draw parallels between metabolic changes observed in cancers or induced by oncoviruses, with a focus on pathways involved in the regulation of glucose, lipid, and amino acids. We describe whether and how oncoviruses depend on metabolic changes, with the perspective of targeting them for antiviral and onco-therapeutic approaches in the context of viral infections.
Collapse
|
64
|
Canciello A, Cerveró-Varona A, Peserico A, Mauro A, Russo V, Morrione A, Giordano A, Barboni B. "In medio stat virtus": Insights into hybrid E/M phenotype attitudes. Front Cell Dev Biol 2022; 10:1038841. [PMID: 36467417 PMCID: PMC9715750 DOI: 10.3389/fcell.2022.1038841] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 11/02/2022] [Indexed: 08/22/2023] Open
Abstract
Epithelial-mesenchymal plasticity (EMP) refers to the ability of cells to dynamically interconvert between epithelial (E) and mesenchymal (M) phenotypes, thus generating an array of hybrid E/M intermediates with mixed E and M features. Recent findings have demonstrated how these hybrid E/M rather than fully M cells play key roles in most of physiological and pathological processes involving EMT. To this regard, the onset of hybrid E/M state coincides with the highest stemness gene expression and is involved in differentiation of either normal and cancer stem cells. Moreover, hybrid E/M cells are responsible for wound healing and create a favorable immunosuppressive environment for tissue regeneration. Nevertheless, hybrid state is responsible of metastatic process and of the increasing of survival, apoptosis and therapy resistance in cancer cells. The present review aims to describe the main features and the emerging concepts regulating EMP and the formation of E/M hybrid intermediates by describing differences and similarities between cancer and normal hybrid stem cells. In particular, the comprehension of hybrid E/M cells biology will surely advance our understanding of their features and how they could be exploited to improve tissue regeneration and repair.
Collapse
Affiliation(s)
- Angelo Canciello
- Faculty of Bioscience and Technology for Food Agriculture and Environment, University of Teramo, Teramo, Italy
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA, United States
| | - Adrián Cerveró-Varona
- Faculty of Bioscience and Technology for Food Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Alessia Peserico
- Faculty of Bioscience and Technology for Food Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Annunziata Mauro
- Faculty of Bioscience and Technology for Food Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Valentina Russo
- Faculty of Bioscience and Technology for Food Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Andrea Morrione
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA, United States
| | - Antonio Giordano
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA, United States
- Sbarro Health Research Organization (SHRO), Philadelphia, PA, United States
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Barbara Barboni
- Faculty of Bioscience and Technology for Food Agriculture and Environment, University of Teramo, Teramo, Italy
| |
Collapse
|
65
|
Liu R, Liu X. Virtual Screening and Biological Activity Evaluation of New Potent Inhibitors Targeting Hexokinase-II. Molecules 2022; 27:7555. [PMID: 36364382 PMCID: PMC9658052 DOI: 10.3390/molecules27217555] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/30/2022] [Accepted: 11/02/2022] [Indexed: 09/29/2023] Open
Abstract
Hexokinase-II (HK-II), the rate-limiting step enzyme in the glycolysis pathway, expresses high levels of cancer cells compared with normal cells. Due to its pivotal role in the different aspects of cancer physiology including cellular proliferation, metastasis, and apoptosis, HK-II provides a new therapeutic target for cancer therapy. The structure-based virtual screening targeting HK-II was used to hit identifications from small molecule databases, and the select compounds were further evaluated in biological assays. Forty-seven compounds with the lowest binding energies were identified as potential HK-II inhibitors. Among them, nine compounds displayed the highest cytotoxicity to three different cancer cells. Based on the mechanism study, compounds 4244-3659 and K611-0094 showed an obvious inhibitory effect on the HK-II enzyme. This study identified two potential inhibitors of HK-II and can be helpful for developing potential drugs targeting HK-II in tumor therapy.
Collapse
Affiliation(s)
- Ruijuan Liu
- College of Physical Education, Northwest Normal University, Lanzhou 730070, China
| | - Xuewei Liu
- Department of Pharmacy, Jiangsu Food and Pharmaceutical Science College, Huaian 223003, China
| |
Collapse
|
66
|
Moss DY, McCann C, Kerr EM. Rerouting the drug response: Overcoming metabolic adaptation in KRAS-mutant cancers. Sci Signal 2022; 15:eabj3490. [PMID: 36256706 DOI: 10.1126/scisignal.abj3490] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Mutations in guanosine triphosphatase KRAS are common in lung, colorectal, and pancreatic cancers. The constitutive activity of mutant KRAS and its downstream signaling pathways induces metabolic rewiring in tumor cells that can promote resistance to existing therapeutics. In this review, we discuss the metabolic pathways that are altered in response to treatment and those that can, in turn, alter treatment efficacy, as well as the role of metabolism in the tumor microenvironment (TME) in dictating the therapeutic response in KRAS-driven cancers. We highlight metabolic targets that may provide clinical opportunities to overcome therapeutic resistance and improve survival in patients with these aggressive cancers.
Collapse
Affiliation(s)
- Deborah Y Moss
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, BT9 7AE Northern Ireland, UK
| | - Christopher McCann
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, BT9 7AE Northern Ireland, UK
| | - Emma M Kerr
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, BT9 7AE Northern Ireland, UK
| |
Collapse
|
67
|
Hansen GE, Gibson GE. The α-Ketoglutarate Dehydrogenase Complex as a Hub of Plasticity in Neurodegeneration and Regeneration. Int J Mol Sci 2022; 23:12403. [PMID: 36293260 PMCID: PMC9603878 DOI: 10.3390/ijms232012403] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/06/2022] [Accepted: 10/11/2022] [Indexed: 07/30/2023] Open
Abstract
Abnormal glucose metabolism is central to neurodegeneration, and considerable evidence suggests that abnormalities in key enzymes of the tricarboxylic acid (TCA) cycle underlie the metabolic deficits. Significant recent advances in the role of metabolism in cancer provide new insight that facilitates our understanding of the role of metabolism in neurodegeneration. Research indicates that the rate-limiting step of the TCA cycle, the α-ketoglutarate dehydrogenase complex (KGDHC) and its substrate alpha ketoglutarate (KG), serve as a signaling hub that regulates multiple cellular processes: (1) is the rate-limiting step of the TCA cycle, (2) is sensitive to reactive oxygen species (ROS) and produces ROS, (3) determines whether KG is used for energy or synthesis of compounds to support growth, (4) regulates the cellular responses to hypoxia, (5) controls the post-translational modification of hundreds of cell proteins in the mitochondria, cytosol, and nucleus through succinylation, (6) controls critical aspects of transcription, (7) modulates protein signaling within cells, and (8) modulates cellular calcium. The primary focus of this review is to understand how reductions in KGDHC are translated to pathologically important changes that underlie both neurodegeneration and cancer. An understanding of each role is necessary to develop new therapeutic strategies to treat neurodegenerative disease.
Collapse
Affiliation(s)
- Grace E. Hansen
- Department of Biology, University of Massachusetts, Lowell, MA 01852, USA
| | - Gary E. Gibson
- Weill Cornell Medicine, Brain and Mind Research Institute, Burke Neurological Institute, White Plains, NY 10605, USA
| |
Collapse
|
68
|
Zhang B, Thorne RF, Zhang P, Wu M, Liu L. Vanguard is a Glucose Deprivation-Responsive Long Non-Coding RNA Essential for Chromatin Remodeling-Reliant DNA Repair. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2201210. [PMID: 36047643 PMCID: PMC9596831 DOI: 10.1002/advs.202201210] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 07/29/2022] [Indexed: 06/15/2023]
Abstract
Glucose metabolism contributes to DNA damage response pathways by regulating chromatin remodeling, double-strand break (DSB) repair, and redox homeostasis, although the underlying mechanisms are not fully established. Here, a previously uncharacterized long non-coding RNA is revealed that is call Vanguard which acts to promote HMGB1-dependent DNA repair in association with changes in global chromatin accessibility. Vanguard expression is maintained in cancer cells by SP1-dependent transcription according to glucose availability and cellular adenosine triphosphate (ATP) levels. Vanguard promotes complex formation between HMGB1 and HDAC1, with the resulting deacetylation of HMGB1 serving to maintain its nuclear localization and DSB repair function. However, Vanguard downregulation under glucose limiting conditions promotes HMGB1 translocation from the nucleus, increasing DNA damage, and compromising cancer cell growth and viability. Moreover, Vanguard silencing increases the effectiveness of poly (ADP-ribose) polymerase inhibitors against breast cancer cells with wild-type breast cancer gene-1 status, suggesting Vanguard as a potential therapeutic target.
Collapse
Affiliation(s)
- Ben Zhang
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230001China
| | - Rick Francis Thorne
- Henan Provincial and Zhengzhou City Key laboratory of Non‐coding RNA and Cancer MetabolismHenan International Join Laboratory of Non‐coding RNA and Metabolism in CancerPeople's Hospital of Zhengzhou UniversityAcademy of Medical SciencesZhengzhou UniversityZhengzhouHenan450053China
| | - Pengfei Zhang
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230001China
- The Cancer Hospital of the University of Chinese Academy of SciencesInstitute of Basic Medicine and Cancer (IBMC)Chinese Academy of SciencesHangzhouZhejiang310022China
| | - Mian Wu
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230001China
- Henan Provincial and Zhengzhou City Key laboratory of Non‐coding RNA and Cancer MetabolismHenan International Join Laboratory of Non‐coding RNA and Metabolism in CancerPeople's Hospital of Zhengzhou UniversityAcademy of Medical SciencesZhengzhou UniversityZhengzhouHenan450053China
| | - Lianxin Liu
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230001China
- Anhui Province Key Laboratory of Hepatopancreatobiliary SurgeryThe First Affiliated Hospital of USTCHefeiAnhui230001China
- Anhui Provincial Clinical Research Center for Hepatobiliary DiseasesThe First Affiliated Hospital of USTCHefeiAnhui230001China
| |
Collapse
|
69
|
Oudaert I, Van der Vreken A, Maes A, De Bruyne E, De Veirman K, Vanderkerken K, Menu E. Metabolic cross-talk within the bone marrow milieu: focus on multiple myeloma. Exp Hematol Oncol 2022; 11:49. [PMID: 36050788 PMCID: PMC9438316 DOI: 10.1186/s40164-022-00303-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/23/2022] [Indexed: 11/17/2022] Open
Abstract
Cancer cells are well-known for their capacity to adapt their metabolism to their increasing energy demands which is necessary for tumor progression. This is no different for Multiple Myeloma (MM), a hematological cancer which develops in the bone marrow (BM), whereby the malignant plasma cells accumulate and impair normal BM functions. It has become clear that the hypoxic BM environment contributes to metabolic rewiring of the MM cells, including changes in metabolite levels, increased/decreased activity of metabolic enzymes and metabolic shifts. These adaptations will lead to a pro-tumoral environment stimulating MM growth and drug resistance In this review, we discuss the identified metabolic changes in MM and the BM microenvironment and summarize how these identified changes have been targeted (by inhibitors, genetic approaches or deprivation studies) in order to block MM progression and survival.
Collapse
Affiliation(s)
- Inge Oudaert
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Arne Van der Vreken
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Anke Maes
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Elke De Bruyne
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Kim De Veirman
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Karin Vanderkerken
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Eline Menu
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090, Brussels, Belgium.
| |
Collapse
|
70
|
Seyfried TN, Arismendi-Morillo G, Zuccoli G, Lee DC, Duraj T, Elsakka AM, Maroon JC, Mukherjee P, Ta L, Shelton L, D'Agostino D, Kiebish M, Chinopoulos C. Metabolic management of microenvironment acidity in glioblastoma. Front Oncol 2022; 12:968351. [PMID: 36059707 PMCID: PMC9428719 DOI: 10.3389/fonc.2022.968351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/15/2022] [Indexed: 11/24/2022] Open
Abstract
Glioblastoma (GBM), similar to most cancers, is dependent on fermentation metabolism for the synthesis of biomass and energy (ATP) regardless of the cellular or genetic heterogeneity seen within the tumor. The transition from respiration to fermentation arises from the documented defects in the number, the structure, and the function of mitochondria and mitochondrial-associated membranes in GBM tissue. Glucose and glutamine are the major fermentable fuels that drive GBM growth. The major waste products of GBM cell fermentation (lactic acid, glutamic acid, and succinic acid) will acidify the microenvironment and are largely responsible for drug resistance, enhanced invasion, immunosuppression, and metastasis. Besides surgical debulking, therapies used for GBM management (radiation, chemotherapy, and steroids) enhance microenvironment acidification and, although often providing a time-limited disease control, will thus favor tumor recurrence and complications. The simultaneous restriction of glucose and glutamine, while elevating non-fermentable, anti-inflammatory ketone bodies, can help restore the pH balance of the microenvironment while, at the same time, providing a non-toxic therapeutic strategy for killing most of the neoplastic cells.
Collapse
Affiliation(s)
- Thomas N. Seyfried
- Biology Department, Boston College, Chestnut Hill, MA, United States
- *Correspondence: Thomas N. Seyfried,
| | - Gabriel Arismendi-Morillo
- Instituto de Investigaciones Biológicas, Facultad de Medicina, Universidad del Zulia, Maracaibo, Venezuela
| | - Giulio Zuccoli
- The Program for the Study of Neurodevelopment in Rare Disorders (NDRD), University of Pittsburgh, Pittsburgh, PA, United States
| | - Derek C. Lee
- Biology Department, Boston College, Chestnut Hill, MA, United States
| | - Tomas Duraj
- Faculty of Medicine, Institute for Applied Molecular Medicine (IMMA), CEU San Pablo University, Madrid, Spain
| | - Ahmed M. Elsakka
- Neuro Metabolism, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Joseph C. Maroon
- Department of Neurosurgery, University of Pittsburgh, Medical Center, Pittsburgh, PA, United States
| | - Purna Mukherjee
- Biology Department, Boston College, Chestnut Hill, MA, United States
| | - Linh Ta
- Biology Department, Boston College, Chestnut Hill, MA, United States
| | | | - Dominic D'Agostino
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, United States
| | | | | |
Collapse
|
71
|
Metabolic targeting of malignant tumors: a need for systemic approach. J Cancer Res Clin Oncol 2022; 149:2115-2138. [PMID: 35925428 DOI: 10.1007/s00432-022-04212-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 07/14/2022] [Indexed: 12/09/2022]
Abstract
PURPOSE Dysregulated metabolism is now recognized as a fundamental hallmark of carcinogenesis inducing aggressive features and additional hallmarks. In this review, well-established metabolic changes displayed by tumors are highlighted in a comprehensive manner and corresponding therapeutical targets are discussed to set up a framework for integrating basic research findings with clinical translation in oncology setting. METHODS Recent manuscripts of high research impact and relevant to the field from PubMed (2000-2021) have been reviewed for this article. RESULTS Metabolic pathway disruption during tumor evolution is a dynamic process potentiating cell survival, dormancy, proliferation and invasion even under dismal conditions. Apart from cancer cells, though, tumor microenvironment has an acting role as extracellular metabolites, pH alterations and stromal cells reciprocally interact with malignant cells, ultimately dictating tumor-promoting responses, disabling anti-tumor immunity and promoting resistance to treatments. CONCLUSION In the field of cancer metabolism, there are several emerging prognostic and therapeutic targets either in the form of gene expression, enzyme activity or metabolites which could be exploited for clinical purposes; both standard-of-care and novel treatments may be evaluated in the context of metabolism rewiring and indeed, synergistic effects between metabolism-targeting and other therapies would be an attractive perspective for further research.
Collapse
|
72
|
Lee H, Woo SM, Jang H, Kang M, Kim SY. Cancer depends on fatty acids for ATP production: A possible link between cancer and obesity. Semin Cancer Biol 2022; 86:347-357. [PMID: 35868515 DOI: 10.1016/j.semcancer.2022.07.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 07/15/2022] [Accepted: 07/16/2022] [Indexed: 12/14/2022]
Abstract
Several metabolic pathways for the supply of adenosine triphosphate (ATP) have been proposed; however, the major source of reducing power for ADP in cancer remains unclear. Although glycolysis is the source of ATP in tumors according to the Warburg effect, ATP levels do not differ between cancer cells grown in the presence and absence of glucose. Several theories have been proposed to explain the supply of ATP in cancer, including metabolic reprograming in the tumor microenvironment. However, these theories are based on the production of ATP by the TCA-OxPhos pathway, which is inconsistent with the Warburg effect. We found that blocking fatty acid oxidation (FAO) in the presence of glucose significantly decreased ATP production in various cancer cells. This suggests that cancer cells depend on fatty acids to produce ATP through FAO instead of glycolysis. We observed that cancer cell growth mainly relies on metabolic nutrients and oxygen systemically supplied through the bloodstream instead of metabolic reprogramming. In a spontaneous mouse tumor model (KrasG12D; Pdx1-cre), tumor growth was 2-fold higher in mice fed a high-fat diet (low-carbo diet) that caused obesity, whereas a calorie-balanced, low-fat diet (high-carbo diet) inhibited tumor growth by 3-fold compared with that in mice fed a control/normal diet. This 5-fold difference in tumor growth between mice fed low-fat and high-fat diets suggests that fat-induced obesity promotes cancer growth, and tumor growth depends on fatty acids as the primary source of energy.
Collapse
Affiliation(s)
- Ho Lee
- Division of Cancer Biology, Research Institute, National Cancer Center, Goyang, Gyeonggi-do 10408, Republic of Korea; Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Gyeonggi-do 10408, Republic of Korea
| | - Sang Myung Woo
- Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Gyeonggi-do 10408, Republic of Korea; Center for Liver and Pancreatobiliary Cancer, National Cancer Center, Goyang, Gyeonggi-do 10408, Republic of Korea
| | - Hyonchol Jang
- Division of Cancer Biology, Research Institute, National Cancer Center, Goyang, Gyeonggi-do 10408, Republic of Korea; Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Gyeonggi-do 10408, Republic of Korea
| | - Mingyu Kang
- Division of Cancer Biology, Research Institute, National Cancer Center, Goyang, Gyeonggi-do 10408, Republic of Korea; New Cancer Cure-Bio Co., Goyang, Gyeonggi-do 10408, Republic of Korea
| | - Soo-Youl Kim
- Division of Cancer Biology, Research Institute, National Cancer Center, Goyang, Gyeonggi-do 10408, Republic of Korea; New Cancer Cure-Bio Co., Goyang, Gyeonggi-do 10408, Republic of Korea.
| |
Collapse
|
73
|
El-Gammal Z, Nasr MA, Elmehrath AO, Salah RA, Saad SM, El-Badri N. Regulation of mitochondrial temperature in health and disease. Pflugers Arch 2022; 474:1043-1051. [PMID: 35780250 PMCID: PMC9492600 DOI: 10.1007/s00424-022-02719-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 06/15/2022] [Indexed: 11/30/2022]
Abstract
Mitochondrial temperature is produced by various metabolic processes inside the mitochondria, particularly oxidative phosphorylation. It was recently reported that mitochondria could normally operate at high temperatures that can reach 50℃. The aim of this review is to identify mitochondrial temperature differences between normal cells and cancer cells. Herein, we discussed the different types of mitochondrial thermosensors and their advantages and disadvantages. We reviewed the studies assessing the mitochondrial temperature in cancer cells and normal cells. We shed the light on the factors involved in maintaining the mitochondrial temperature of normal cells compared to cancer cells.
Collapse
Affiliation(s)
- Zaynab El-Gammal
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, Cairo, Egypt.,Egypt Center for Research and Regenerative Medicine, Cairo, Egypt
| | - Mohamed A Nasr
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, Cairo, Egypt
| | - Ahmed O Elmehrath
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, Cairo, Egypt.,Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Radwa A Salah
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, Cairo, Egypt
| | - Shams M Saad
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, Cairo, Egypt
| | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, Cairo, Egypt.
| |
Collapse
|
74
|
Ohno O, Iwasaki A, Same K, Kudo C, Aida E, Sugiura K, Sumimoto S, Teruya T, Tashiro E, Simizu S, Matsuno K, Imoto M, Suenaga K. Isolation of Caldorazole, a Thiazole-Containing Polyketide with Selective Cytotoxicity under Glucose-Restricted Conditions. Org Lett 2022; 24:4547-4551. [PMID: 35713373 DOI: 10.1021/acs.orglett.2c01566] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Caldorazole (1) was isolated from the marine cyanobacterium Caldora sp. collected on Ishigaki Island, Okinawa, Japan. Its structure was determined to be a new polyketide that contained two thiazole rings and an O-methylenolpyruvamide moiety. Caldorazole (1) showed strong cytotoxicity toward tumor cells that had been seeded at a high density. Cell death induced by 1 in HeLa and A431 cells was also observed only in the presence of the glycolysis blocker 2-deoxy-d-glucose (2DG). Co-treatment with 1 and 2DG remarkably decreased ATP levels in these cells. Furthermore, 1 selectively inhibited complex I in the mitochondrial respiratory chain. Thus, 1 was demonstrated to exert cytotoxicity toward human tumor cells by blocking mitochondrial respiration.
Collapse
Affiliation(s)
- Osamu Ohno
- Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, 2665-1 Nakano, Hachioji 192-0015, Japan
| | | | | | | | - Erika Aida
- Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, 2665-1 Nakano, Hachioji 192-0015, Japan
| | - Kazuya Sugiura
- Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, 2665-1 Nakano, Hachioji 192-0015, Japan
| | | | - Toshiaki Teruya
- Faculty of Education, University of the Ryukyus, 1 Senbaru, Nishihara, Okinawa 903-0213, Japan
| | | | | | - Kenji Matsuno
- Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, 2665-1 Nakano, Hachioji 192-0015, Japan
| | | | | |
Collapse
|
75
|
Vulczak A, Alberici LC. Physical Exercise and Tumor Energy Metabolism. Cancer Treat Res Commun 2022; 32:100600. [PMID: 35811248 DOI: 10.1016/j.ctarc.2022.100600] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 06/25/2022] [Accepted: 06/27/2022] [Indexed: 12/15/2022]
Abstract
Evidence supports the antitumoral effects of physical activity, either in experimental animal models or humans. However, the biological mechanisms by which physical exercise modulates tumoral development are still unclear. An important feature of the tumor cells is the altered energy metabolism, often associated with definitions of tumor aggressiveness. Nevertheless, exercise can cause global metabolic changes in the body, as well as modulate tumor metabolism. Here we specifically discuss the metabolic changes found in tumors and how exercise can contribute to anti-tumoral effects by modulating the mitochondrial function, and tricarboxylic acid cycle-related metabolites of cancer cells. The effect of physical exercise on tumor metabolism is a new possibility for comprehension of cancer biology and developing therapies focused on tumor energy metabolism.
Collapse
Affiliation(s)
- Anderson Vulczak
- Department of Biomolecular Sciences - School of Pharmaceutical Sciences of Ribeirao Preto - University of Sao Paulo, RibeirãoPreto, SP, Brazil
| | - Luciane Carla Alberici
- Department of Biomolecular Sciences - School of Pharmaceutical Sciences of Ribeirao Preto - University of Sao Paulo, RibeirãoPreto, SP, Brazil.
| |
Collapse
|
76
|
Bao S, Wang X, Li M, Gao Z, Zheng D, Shen D, Liu L. Potential of Mitochondrial Ribosomal Genes as Cancer Biomarkers Demonstrated by Bioinformatics Results. Front Oncol 2022; 12:835549. [PMID: 35719986 PMCID: PMC9204274 DOI: 10.3389/fonc.2022.835549] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 04/27/2022] [Indexed: 12/15/2022] Open
Abstract
Next-generation sequencing and bioinformatics analyses have clearly revealed the roles of mitochondrial ribosomal genes in cancer development. Mitochondrial ribosomes are composed of three RNA components encoded by mitochondrial DNA and 82 specific protein components encoded by nuclear DNA. They synthesize mitochondrial inner membrane oxidative phosphorylation (OXPHOS)-related proteins and participate in various biological activities via the regulation of energy metabolism and apoptosis. Mitochondrial ribosomal genes are strongly associated with clinical features such as prognosis and foci metastasis in patients with cancer. Accordingly, mitochondrial ribosomes have become an important focus of cancer research. We review recent advances in bioinformatics research that have explored the link between mitochondrial ribosomes and cancer, with a focus on the potential of mitochondrial ribosomal genes as biomarkers in cancer.
Collapse
Affiliation(s)
- Shunchao Bao
- Department of Radiotherapy, Second Hospital of Jilin University, Changchun, China
| | - Xinyu Wang
- Department of Breast Surgery, Second Hospital of Jilin University, Changchun, China
| | - Mo Li
- Department of Radiotherapy, Second Hospital of Jilin University, Changchun, China
| | - Zhao Gao
- Nuclear Medicine Department, Second Hospital of Jilin University, Changchun, China
| | - Dongdong Zheng
- Department of Cardiovascular Surgery, Second Hospital of Jilin University, Changchun, China
| | - Dihan Shen
- Medical Research Center, Second Hospital of Jilin University, Changchun, China
| | - Linlin Liu
- Department of Radiotherapy, Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
77
|
Gao W, Zhang J, Wang W, Liu Z, Chen M, Hu X, Zeng L, Zheng C, Song H, Zhang Q. Drug Self-delivery Nanorods Enhance Photodynamic Therapy of Triple-Negative Breast Cancer by inhibiting Oxidative Phosphorylation. Int J Pharm 2022; 621:121775. [PMID: 35489603 DOI: 10.1016/j.ijpharm.2022.121775] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 04/08/2022] [Accepted: 04/23/2022] [Indexed: 11/26/2022]
Abstract
Photodynamic therapy (PDT) shows very high potential for the clinical treatment of triple-negative breast cancer. However, the efficacy of PDT is significantly weakened by tumor hypoxia, the relatively high intracellular glutathione levels and the active proliferation of cancer cells. To address these issues, we developed a novel drug self-delivery nanorod (defined as AINRs) through the hydrophobic interaction among the mitochondrial complex III inhibitor (atovaquone, ATO), the photosensitizer (indocyanine green, ICG) and the dispersion stabilizer (distearoyl phosphoethanolamine-polyethylene glycol 2000, DSPE-PEG 2000). The AINRs showed a rod-like morphology with a mean diameter of 120.6 ± 5.4 nm, a zeta potential of -26.35 ± 1.63 mV and a significantly high drug loading rate of 93.48%. The results of in vitro cell experiments involving triple-negative breast cancer cell lines (4T1 cells and MDA-MB-231 cells) indicated that the AINRs could effectively block the oxidative phosphorylation of cancer cells through the inhibition of mitochondrial complex III, which results in the reduction of endogenous oxygen consumption and the decrease of the intracellular ATP level. The reduction of ATP content further inhibited the glutathione synthesis and arrested the cell cycle at the S-phase, which results in enhanced in vitro PDT efficacy of ICG. The results of in vivo antitumor activity in 4T1-bearing mice showed that the tumor growth inhibition rate of the AINRs with near-infrared laser irradiation (NIR) was 90%, whereas the tumor growth inhibition rates of the AINRs without NIR, ICG with NIR and doxorubicin (3 mg/kg) were only 31.68%, 61.15% and 24.59%, respectively. In addition, the results of safety studies, including body weights, biochemical indicators and H&E staining images of the main organs demonstrated the security of the AINRs. In summary, this study showed that the oxidative phosphorylation inhibition of triple-negative breast cancer was a safe and effective method to enhance its PDT efficacy.
Collapse
Affiliation(s)
- Wenhao Gao
- College of Pharmacy, Fujian Medical University, Fuzhou 350122, PR China; Department of Pharmacy, Fuzong Clinical Medical College of Fujian Medical University (900 Hospital of the Joint Logistics Team), Fuzhou 350025, PR China
| | - Jialiang Zhang
- Innovation center for cancer research, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou 350014, PR China
| | - Weifeng Wang
- College of Pharmacy, Fujian Medical University, Fuzhou 350122, PR China; Department of Pharmacy, Fuzong Clinical Medical College of Fujian Medical University (900 Hospital of the Joint Logistics Team), Fuzhou 350025, PR China
| | - Zhihong Liu
- Department of Pharmacy, Fuzong Clinical Medical College of Fujian Medical University (900 Hospital of the Joint Logistics Team), Fuzhou 350025, PR China
| | - Mulan Chen
- Department of Breast Cancer, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou 350014, PR China
| | - Xiaomu Hu
- Department of Pharmacy, Fuzong Clinical Medical College of Fujian Medical University (900 Hospital of the Joint Logistics Team), Fuzhou 350025, PR China
| | - Lingjun Zeng
- Department of Pharmacy, Fuzong Clinical Medical College of Fujian Medical University (900 Hospital of the Joint Logistics Team), Fuzhou 350025, PR China
| | - Changqing Zheng
- Department of Pharmacy, Fuzong Clinical Medical College of Fujian Medical University (900 Hospital of the Joint Logistics Team), Fuzhou 350025, PR China
| | - Hongtao Song
- College of Pharmacy, Fujian Medical University, Fuzhou 350122, PR China; Department of Pharmacy, Fuzong Clinical Medical College of Fujian Medical University (900 Hospital of the Joint Logistics Team), Fuzhou 350025, PR China.
| | - Qian Zhang
- College of Pharmacy, Fujian Medical University, Fuzhou 350122, PR China.
| |
Collapse
|
78
|
Kulbay M, Johnson B, Ricaud G, Séguin-Grignon MN, Bernier J. Energetic metabolic reprogramming in Jurkat DFF40-deficient cancer cells. Mol Cell Biochem 2022; 477:2213-2233. [PMID: 35460011 DOI: 10.1007/s11010-022-04433-0] [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: 10/02/2021] [Accepted: 04/04/2022] [Indexed: 11/25/2022]
Abstract
DNA fragmentation factor 40 (DFF40), or the caspase-activated DNase (CAD), is an endonuclease specific for double-stranded DNA. Alterations in its function and expression have been linked to apoptosis resistance, a mechanism likely used by cancer cells. However, how the DFF40-related apoptosis resistance pathway occurs remains unclear. Here, we sought to determine if DFF40 expression could be linked to cell metabolism through the regulation of mitochondrial integrity and function. We demonstrated that DFF40-deficient cells are more resistant to staurosporine and tributyltin (TBT)-induced apoptosis, and express higher levels of Mcl-1 at basal state. Treatment with TBT induces higher Bcl-2 and caspase-9 mRNA transcripts in DFF40 KO Jurkat cells, as well as enhanced Bcl-2 phosphorylation. A loss of DFF40 expression induces a higher mitochondrial mass, mtDNA copy number, mitochondrial membrane potential, and glycolysis rates in resting T cells. DFF40-deficient cells exhibit the Warburg effect phenotype, where they rely significantly more on glycolysis than oxidative phosphorylation and have a higher proliferative state, demonstrated by a higher Ki-67 transcription factor expression and AKT phosphorylation. Finally, we demonstrated with cell fractioning that DFF40 can translocate to the mitochondria following apoptosis induction. Our study reveals that DFF40 may act as a regulator of mitochondria during cell death and its loss could compromise mitochondrial integrity and cause an energetic reprogramming in pathologies such as cancer.
Collapse
Affiliation(s)
- Merve Kulbay
- INRS-Centre Armand-Frappier Santé Biotechnologie, 531 Boul. des Prairies, Laval, QC, H7V 1B7, Canada
- Department of Medicine, Université de Montréal, 2900 Blvd. Edouard Montpetit, Montréal, QC, Canada
| | - Bruno Johnson
- INRS-Centre Armand-Frappier Santé Biotechnologie, 531 Boul. des Prairies, Laval, QC, H7V 1B7, Canada
| | - Guillaume Ricaud
- INRS-Centre Armand-Frappier Santé Biotechnologie, 531 Boul. des Prairies, Laval, QC, H7V 1B7, Canada
| | | | - Jacques Bernier
- INRS-Centre Armand-Frappier Santé Biotechnologie, 531 Boul. des Prairies, Laval, QC, H7V 1B7, Canada.
| |
Collapse
|
79
|
Sabater B. Entropy Perspectives of Molecular and Evolutionary Biology. Int J Mol Sci 2022; 23:ijms23084098. [PMID: 35456917 PMCID: PMC9029946 DOI: 10.3390/ijms23084098] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/01/2022] [Accepted: 04/06/2022] [Indexed: 02/01/2023] Open
Abstract
Attempts to find and quantify the supposed low entropy of organisms and its preservation are revised. The absolute entropy of the mixed components of non-living biomass (approximately −1.6 × 103 J K−1 L−1) is the reference to which other entropy decreases would be ascribed to life. The compartmentation of metabolites and the departure from the equilibrium of metabolic reactions account for reductions in entropy of 1 and 40–50 J K−1 L−1, respectively, and, though small, are distinctive features of living tissues. DNA and proteins do not supply significant decreases in thermodynamic entropy, but their low informational entropy is relevant for life and its evolution. No other living feature contributes significantly to the low entropy associated with life. The photosynthetic conversion of radiant energy to biomass energy accounts for most entropy (2.8 × 105 J K−1 carbon kg−1) produced by living beings. The comparatively very low entropy produced in other processes (approximately 4.8 × 102 J K−1 L−1 day−1 in the human body) must be rapidly exported outside as heat to preserve low entropy decreases due to compartmentation and non-equilibrium metabolism. Enzymes and genes are described, whose control minimizes the rate of production of entropy and could explain selective pressures in biological evolution and the rapid proliferation of cancer cells.
Collapse
Affiliation(s)
- Bartolomé Sabater
- Department of Life Sciences, University of Alcalá, 28805 Alcalá de Henares, Madrid, Spain
| |
Collapse
|
80
|
Drp1-Mediated Mitochondrial Metabolic Dysfunction Inhibits the Tumor Growth of Pituitary Adenomas. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5652586. [PMID: 35368865 PMCID: PMC8967574 DOI: 10.1155/2022/5652586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 12/01/2021] [Accepted: 02/10/2022] [Indexed: 12/04/2022]
Abstract
Metabolic changes have been suggested to be a hallmark of tumors and are closely associated with tumorigenesis. In a previous study, we demonstrated the role of lactate dehydrogenase in regulating abnormal glucose metabolism in pituitary adenomas (PA). As the key organelle of oxidative phosphorylation (OXPHOS), mitochondria play a vital role in the energy supply for tumor cells. However, few attempts have been made to elucidate mitochondrial metabolic homeostasis in PA. Dynamin-related protein 1 (Drp1) is a member of the dynamin superfamily of GTPases, which mediates mitochondrial fission. This study is aimed at investigating whether Drp1 affects the progression of PA through abnormal mitochondrial metabolism. We analyzed the expression of dynamin-related protein 1 (Drp1) in 20 surgical PA samples. The effects of Drp1 on PA growth were assessed in vitro and in xenograft models. We found an upregulation of Drp1 in PA samples with a low proliferation index. Knockdown or inhibition of Drp1 enhanced the proliferation of PA cell lines in vitro, while overexpression of Drp1 could reversed such effects. Mechanistically, overexpressed Drp1 damaged mitochondria by overproduction of reactive oxygen species (ROS), which induced mitochondrial OXPHOS inhibition and decline of ATP production. The energy deficiency inhibited proliferation of PA cells. In addition, overexpressed Drp1 promoted cytochrome c release from damaged mitochondria into the cytoplasm and then activated the downstream caspase apoptotic cascade reaction, which induced apoptosis of PA cells. Moreover, the decreased ATP production induced by Drp1 overexpressing activated the AMPK cellular energy stress sensor and enhanced autophagy through the AMPK-ULK1 pathway, which might play a protective role in PA growth. Furthermore, overexpression of Drp1 repressed PA growth in vivo. Our data indicates that Drp1-mediated mitochondrial metabolic dysfunction inhibits PA growth by affecting cell proliferation, apoptosis, and autophagy. Selectively targeting mitochondrial metabolic homeostasis stands out as a promising antineoplastic strategy for PA therapy.
Collapse
|
81
|
Chemotherapy Resistance: Role of Mitochondrial and Autophagic Components. Cancers (Basel) 2022; 14:cancers14061462. [PMID: 35326612 PMCID: PMC8945922 DOI: 10.3390/cancers14061462] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/10/2022] [Accepted: 03/10/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Chemotherapy resistance is a common occurrence during cancer treatment that cancer researchers are attempting to understand and overcome. Mitochondria are a crucial intracellular signaling core that are becoming important determinants of numerous aspects of cancer genesis and progression, such as metabolic reprogramming, metastatic capability, and chemotherapeutic resistance. Mitophagy, or selective autophagy of mitochondria, can influence both the efficacy of tumor chemotherapy and the degree of drug resistance. Regardless of the fact that mitochondria are well-known for coordinating ATP synthesis from cellular respiration in cellular bioenergetics, little is known its mitophagy regulation in chemoresistance. Recent advancements in mitochondrial research, mitophagy regulatory mechanisms, and their implications for our understanding of chemotherapy resistance are discussed in this review. Abstract Cancer chemotherapy resistance is one of the most critical obstacles in cancer therapy. One of the well-known mechanisms of chemotherapy resistance is the change in the mitochondrial death pathways which occur when cells are under stressful situations, such as chemotherapy. Mitophagy, or mitochondrial selective autophagy, is critical for cell quality control because it can efficiently break down, remove, and recycle defective or damaged mitochondria. As cancer cells use mitophagy to rapidly sweep away damaged mitochondria in order to mediate their own drug resistance, it influences the efficacy of tumor chemotherapy as well as the degree of drug resistance. Yet despite the importance of mitochondria and mitophagy in chemotherapy resistance, little is known about the precise mechanisms involved. As a consequence, identifying potential therapeutic targets by analyzing the signal pathways that govern mitophagy has become a vital research goal. In this paper, we review recent advances in mitochondrial research, mitophagy control mechanisms, and their implications for our understanding of chemotherapy resistance.
Collapse
|
82
|
Ramos JRC, Bissinger T, Genzel Y, Reichl U. Impact of Influenza A Virus Infection on Growth and Metabolism of Suspension MDCK Cells Using a Dynamic Model. Metabolites 2022; 12:metabo12030239. [PMID: 35323683 PMCID: PMC8950586 DOI: 10.3390/metabo12030239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/04/2022] [Accepted: 03/09/2022] [Indexed: 11/21/2022] Open
Abstract
Cell cultured-based influenza virus production is a viable option for vaccine manufacturing. In order to achieve a high concentration of viable cells, is requirement to have not only optimal process conditions, but also an active metabolism capable of intracellular synthesis of viral components. Experimental metabolic data collected in such processes are complex and difficult to interpret, for which mathematical models are an appropriate way to simulate and analyze the complex and dynamic interaction between the virus and its host cell. A dynamic model with 35 states was developed in this study to describe growth, metabolism, and influenza A virus production in shake flask cultivations of suspension Madin-Darby Canine Kidney (MDCK) cells. It considers cell growth (concentration of viable cells, mean cell diameters, volume of viable cells), concentrations of key metabolites both at the intracellular and extracellular level and virus titers. Using one set of parameters, the model accurately simulates the dynamics of mock-infected cells and correctly predicts the overall dynamics of virus-infected cells for up to 60 h post infection (hpi). The model clearly suggests that most changes observed after infection are related to cessation of cell growth and the subsequent transition to apoptosis and cell death. However, predictions do not cover late phases of infection, particularly for the extracellular concentrations of glutamate and ammonium after about 12 hpi. Results obtained from additional in silico studies performed indicated that amino acid degradation by extracellular enzymes resulting from cell lysis during late infection stages may contribute to this observed discrepancy.
Collapse
Affiliation(s)
- João Rodrigues Correia Ramos
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Sandtorstrasse 1, 39106 Magdeburg, Germany; (T.B.); (Y.G.); (U.R.)
- Correspondence:
| | - Thomas Bissinger
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Sandtorstrasse 1, 39106 Magdeburg, Germany; (T.B.); (Y.G.); (U.R.)
| | - Yvonne Genzel
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Sandtorstrasse 1, 39106 Magdeburg, Germany; (T.B.); (Y.G.); (U.R.)
| | - Udo Reichl
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Sandtorstrasse 1, 39106 Magdeburg, Germany; (T.B.); (Y.G.); (U.R.)
- Institute of Process Engineering, Faculty of Process & Systems Engineering, Otto-von-Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany
| |
Collapse
|
83
|
Park JU, Han HJ, Baik JY. Energy metabolism in Chinese hamster ovary (CHO) cells: Productivity and beyond. KOREAN J CHEM ENG 2022. [DOI: 10.1007/s11814-022-1062-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
84
|
Kamada S, Takeiwa T, Ikeda K, Horie K, Inoue S. Emerging Roles of COX7RP and Mitochondrial Oxidative Phosphorylation in Breast Cancer. Front Cell Dev Biol 2022; 10:717881. [PMID: 35178385 PMCID: PMC8844363 DOI: 10.3389/fcell.2022.717881] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 01/17/2022] [Indexed: 12/14/2022] Open
Abstract
Metabolic alterations are critical events in cancers, which often contribute to tumor pathophysiology. While aerobic glycolysis is a known characteristic of cancer-related metabolism, recent studies have shed light on mitochondria-related metabolic pathways in cancer biology, including oxidative phosphorylation (OXPHOS), amino acid and lipid metabolism, nucleic acid metabolism, and redox regulation. Breast cancer is the most common cancer in women; thus, elucidation of breast cancer-related metabolic alteration will help to develop cancer drugs for many patients. We here aim to define the contribution of mitochondrial metabolism to breast cancer biology. The relevance of OXPHOS in breast cancer has been recently defined by the discovery of COX7RP, which promotes mitochondrial respiratory supercomplex assembly and glutamine metabolism: the latter is also shown to promote nucleic acid and fatty acid biosynthesis as well as ROS defense regulation. In this context, the estrogen-related receptor (ERR) family nuclear receptors and collaborating coactivators peroxisome proliferator-activated receptor-γ coactivator-1 (PGC-1) are essential transcriptional regulators for both energy production and cancer-related metabolism. Summarizing recent findings of mitochondrial metabolism in breast cancer, this review will aim to provide a clue for the development of alternative clinical management by modulating the activities of responsible molecules involved in disease-specific metabolic alterations.
Collapse
Affiliation(s)
- Shuhei Kamada
- Division of Systems Medicine and Gene Therapy, Saitama Medical University, Saitama, Japan.,Department of Urology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Toshihiko Takeiwa
- Department of Systems Aging Science and Medicine, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Kazuhiro Ikeda
- Division of Systems Medicine and Gene Therapy, Saitama Medical University, Saitama, Japan
| | - Kuniko Horie
- Division of Systems Medicine and Gene Therapy, Saitama Medical University, Saitama, Japan
| | - Satoshi Inoue
- Division of Systems Medicine and Gene Therapy, Saitama Medical University, Saitama, Japan.,Department of Systems Aging Science and Medicine, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| |
Collapse
|
85
|
Zhang W, Wang C, Hu X, Lian Y, Ding C, Ming L. Inhibition of LDHA suppresses cell proliferation and increases mitochondrial apoptosis via the JNK signaling pathway in cervical cancer cells. Oncol Rep 2022; 47:77. [PMID: 35191522 PMCID: PMC8892607 DOI: 10.3892/or.2022.8288] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 01/28/2022] [Indexed: 11/08/2022] Open
Abstract
The Warburg effect or aerobic glycolysis is a hallmark of cancer. Lactate dehydrogenase (LDH), which catalyzes conversion of pyruvate into lactate, serves a critical role during Warburg effect. LDH A chain (LDHA), a member of the LDH family, is upregulated in multiple types of cancer and serves a vital role in tumor growth and progression. However, its expression and function in cervical cancer has not been characterized. The present study evaluated LDHA expression in The Cancer Genome Atlas database and found that LDHA was upregulated in cervical cancer compared with normal tissue. To clarify the role of LDHA in cervical cancer HeLa and SiHa cells, lentiviral shRNA was used to stably knockdown LDHA and oxamate, a small-molecule inhibitor of LDHA, was used to inhibit the activity of LDHA. Glucose uptake assay, lactate production measurement and ATP detection assay demonstrated LDHA inhibition notably decreased glucose consumption, lactate production and ATP levels in both HeLa and SiHa cells. Furthermore, the effect of LDHA inhibition on cell proliferation, cell cycle and apoptosis was investigated by MTT, BrdU incorporation, colony formation assay, flow cytometry and western blotting; LDHA knockdown or oxamate treatment led to decreased cell proliferation and increased apoptosis. Inhibition of LDHA induced G2/M cell cycle arrest and activated the mitochondrial apoptosis pathway. Mechanistically, the JNK signaling pathway was key for LDHA inhibition-mediated cell cycle arrest and apoptosis. Collectively, these results indicated that LDHA was involved in cervical cancer pathogenesis and may be a promising therapeutic target for treatment.
Collapse
Affiliation(s)
- Wenjing Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R China
| | - Cui Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R China
| | - Xiaomei Hu
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R China
| | - Yanzhen Lian
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R China
| | - Caili Ding
- Zhengzhou Hang Gang Ding Shi Medical Laboratory Co., Ltd., Zhengzhou, Henan 450000, P.R. China
| | - Liang Ming
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R Chin
| |
Collapse
|
86
|
Xiao MH, Lin YF, Xie PP, Chen HX, Deng JW, Zhang W, Zhao N, Xie C, Meng Y, Liu X, Zhuang SM, Zhu Y, Fang JH. Downregulation of a mitochondrial micropeptide, MPM, promotes hepatoma metastasis by enhancing mitochondrial complex I activity. Mol Ther 2022; 30:714-725. [PMID: 34478872 PMCID: PMC8821931 DOI: 10.1016/j.ymthe.2021.08.032] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 07/25/2021] [Accepted: 08/25/2021] [Indexed: 02/07/2023] Open
Abstract
We and others have shown that MPM (micropeptide in mitochondria) regulates myogenic differentiation and muscle development. However, the roles of MPM in cancer development remain unknown. Here we revealed that MPM was downregulated significantly in human hepatocellular carcinoma (HCC) tissues and its decrease was associated with increased metastasis potential and HCC recurrence. Gain- and loss-of-function investigations disclosed that in vitro migration/invasion and in vivo liver/lung metastasis of hepatoma cells were repressed by restoring MPM expression and increased by silencing MPM. Mechanism investigations revealed that MPM interacted with NDUFA7. Mitochondrial complex I activity was inhibited by overexpressing MPM and enhanced by siMPM, and this effect of siMPM was attenuated by knocking down NDUFA7. The NAD+/NADH ratio, which was regulated by complex I, was reduced by MPM but increased by siMPM. Treatment with the NAD+ precursor nicotinamide abrogated the inhibitory effect of MPM on hepatoma cell migration. Further investigations showed that miR-17-5p bound to MPM and inhibited MPM expression. miR-17-5p upregulation was associated with MPM downregulation in HCC tissues. These findings indicate that a decrease in MPM expression may promote hepatoma metastasis by increasing mitochondrial complex I activity and the NAD+/NADH ratio.
Collapse
Affiliation(s)
- Man-Huan Xiao
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Xin Gang Xi Road #135, Guangzhou 510275, P.R. China
| | - Yi-Fang Lin
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Xin Gang Xi Road #135, Guangzhou 510275, P.R. China
| | - Peng-Peng Xie
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Xin Gang Xi Road #135, Guangzhou 510275, P.R. China
| | - Hua-Xing Chen
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Xin Gang Xi Road #135, Guangzhou 510275, P.R. China
| | - Jun-Wen Deng
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Xin Gang Xi Road #135, Guangzhou 510275, P.R. China
| | - Wei Zhang
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Xin Gang Xi Road #135, Guangzhou 510275, P.R. China
| | - Na Zhao
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Xin Gang Xi Road #135, Guangzhou 510275, P.R. China
| | - Chen Xie
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Xin Gang Xi Road #135, Guangzhou 510275, P.R. China
| | - Yu Meng
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Xin Gang Xi Road #135, Guangzhou 510275, P.R. China
| | - Xingguo Liu
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou 510530, China
| | - Shi-Mei Zhuang
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Xin Gang Xi Road #135, Guangzhou 510275, P.R. China,Corresponding author: Shi-Mei Zhuang, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Xin Gang Xi Road #135, Guangzhou 510275, P.R. China.
| | - Ying Zhu
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Xin Gang Xi Road #135, Guangzhou 510275, P.R. China,Corresponding author: Ying Zhu, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Xin Gang Xi Road #135, Guangzhou 510275, P.R. China.
| | - Jian-Hong Fang
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Xin Gang Xi Road #135, Guangzhou 510275, P.R. China,Corresponding author: Jian-Hong Fang, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Xin Gang Xi Road #135, Guangzhou 510275, P.R. China.
| |
Collapse
|
87
|
Bedi M, Ray M, Ghosh A. Active mitochondrial respiration in cancer: a target for the drug. Mol Cell Biochem 2022; 477:345-361. [PMID: 34716860 DOI: 10.1007/s11010-021-04281-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 10/21/2021] [Indexed: 12/21/2022]
Abstract
The relative contribution of mitochondrial respiration and subsequent energy production in malignant cells has remained controversial to date. Enhanced aerobic glycolysis and impaired mitochondrial respiration have gained more attention in the metabolic study of cancer. In contrast to the popular concept, mitochondria of cancer cells oxidize a diverse array of metabolic fuels to generate a majority of the cellular energy by respiration. Several mitochondrial respiratory chain (MRC) subunits' expressions are critical for the growth, metastasis, and cancer cell invasion. Also, the assembly factors, which regulate the integration of individual MRC complexes into native super-complexes, are upregulated in cancer. Moreover, a series of anti-cancer drugs function by inhibiting respiration and ATP production. In this review, we have specified the roles of mitochondrial fuels, MRC subunits, and super-complex assembly factors that promote active respiration across different cancer types and discussed the potential roles of MRC inhibitor drugs in controlling cancer.
Collapse
Affiliation(s)
- Minakshi Bedi
- Department of Biochemistry, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, West Bengal, 700019, India
| | - Manju Ray
- Department of Biophysics, Bose Institute, P 1/12, CIT Scheme VII M, Kolkata, West Bengal, 700054, India
- Department of Chemistry, Institute of Applied Science & Humanities GLA University Mathura, 17km Stone, NH-2, Mathura-Delhi Road, Mathura, UP, 281 406, India
| | - Alok Ghosh
- Department of Biochemistry, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, West Bengal, 700019, India.
| |
Collapse
|
88
|
Wu Q, Wang SP, Sun XX, Tao YF, Yuan XQ, Chen QM, Dai L, Li CL, Zhang JY, Yang AL. HuaChanSu suppresses tumor growth and interferes with glucose metabolism in hepatocellular carcinoma cells by restraining Hexokinase-2. Int J Biochem Cell Biol 2022; 142:106123. [PMID: 34826616 DOI: 10.1016/j.biocel.2021.106123] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/30/2021] [Accepted: 11/17/2021] [Indexed: 02/06/2023]
Abstract
Hepatocellular carcinoma (HCC) has become the sixth highly diagnosed cancer and the fourth main reason of cancer deaths worldwide. HuaChanSu, an extract from dried toad skin, exhibits good anticancer effects and has been widely used in the treatment of liver cancer. The reprogramming of glucose metabolism is one remarkable feature of hepatocellular carcinoma, and the effects of HuaChanSu on the abnormal glucose metabolism of cancer cells have not been elucidated. In our study, we investigate the effects of HuaChanSu on glucose metabolism of hepatocellular carcinoma cells and tumor growth in vivo. The results show that HuaChanSu inhibits the tumor growth of hepatoma H22-bearing mice and prolongs the survival time of tumor-bearing mice, additionally, HuaChanSu has no obvious adverse effects in these mice. In vitro, HuaChanSu restrains the proliferation, induces apoptosis and cell cycle arrest of human hepatoma cells. HuaChanSu also promotes ROS production and causes mitochondrial damage. Furthermore, HuaChanSu inhibits glucose uptake and lactate release in human hepatoma cells. Mechanistically, we find that HuaChanSu downregulates Hexokinase-2 (HK2) expression, and using RNA interference, we confirm that HuaChanSu suppresses the growth of HepG2 cells by interfering with glucose metabolism through downregulation of Hexokinase-2. However, knockdown of Hexokinase-2 has no obvious effect on the proliferation of SK-HEP-1 cells, although glucose uptake and lactate release are reduced in siHK2-transfected SK-HEP-1 cells, subsequently, we illustrate that two human hepatoma cell lines exhibit glucose metabolism heterogeneity, which causes the different cell proliferation responses to the inhibition of Hexokinase-2. Taken together, our study indicates that HuaChanSu could inhibit tumor growth and interfere with glucose metabolism via suppression of Hexokinase-2, and these findings provide a new insight into the anti-hepatoma mechanisms of HuaChanSu and lay a theoretical foundation for the further clinical application of HuaChanSu.
Collapse
Affiliation(s)
- Qi Wu
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Shao-Ping Wang
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Xiao-Xue Sun
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Yu-Fan Tao
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Xiao-Qing Yuan
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Qi-Mei Chen
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Long Dai
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Chun-Lei Li
- Biotechnological Institute of Chinese Materia Medica, Jinan University, Guangzhou 510632, China.
| | - Jia-Yu Zhang
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China.
| | - Ai-Lin Yang
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China.
| |
Collapse
|
89
|
de Beauchamp L, Himonas E, Helgason GV. Mitochondrial metabolism as a potential therapeutic target in myeloid leukaemia. Leukemia 2022; 36:1-12. [PMID: 34561557 PMCID: PMC8727299 DOI: 10.1038/s41375-021-01416-w] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 09/06/2021] [Accepted: 09/07/2021] [Indexed: 02/07/2023]
Abstract
While the understanding of the genomic aberrations that underpin chronic and acute myeloid leukaemia (CML and AML) has allowed the development of therapies for these diseases, limitations remain. These become apparent when looking at the frequency of treatment resistance leading to disease relapse in leukaemia patients. Key questions regarding the fundamental biology of the leukaemic cells, such as their metabolic dependencies, are still unresolved. Even though a majority of leukaemic cells are killed during initial treatment, persistent leukaemic stem cells (LSCs) and therapy-resistant cells are still not eradicated with current treatments, due to various mechanisms that may contribute to therapy resistance, including cellular metabolic adaptations. In fact, recent studies have shown that LSCs and treatment-resistant cells are dependent on mitochondrial metabolism, hence rendering them sensitive to inhibition of mitochondrial oxidative phosphorylation (OXPHOS). As a result, rewired energy metabolism in leukaemic cells is now considered an attractive therapeutic target and the significance of this process is increasingly being recognised in various haematological malignancies. Therefore, identifying and targeting aberrant metabolism in drug-resistant leukaemic cells is an imperative and a relevant strategy for the development of new therapeutic options in leukaemia. In this review, we present a detailed overview of the most recent studies that present experimental evidence on how leukaemic cells can metabolically rewire, more specifically the importance of OXPHOS in LSCs and treatment-resistant cells, and the current drugs available to target this process. We highlight that uncovering specific energy metabolism dependencies will guide the identification of new and more targeted therapeutic strategies for myeloid leukaemia.
Collapse
Affiliation(s)
- Lucie de Beauchamp
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Ekaterini Himonas
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - G Vignir Helgason
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
90
|
Choubey P, Kaur H, Bansal K. Modulation of DNA/RNA Methylation Signaling Mediating Metabolic Homeostasis in Cancer. Subcell Biochem 2022; 100:201-237. [PMID: 36301496 DOI: 10.1007/978-3-031-07634-3_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] [Indexed: 06/16/2023]
Abstract
Nucleic acid methylation is a fundamental epigenetic mechanism that impinges upon several cellular attributes, including metabolism and energy production. The dysregulation of deoxyribonucleic acid (DNA)/ribonucleic acid (RNA) methylation can lead to metabolic rewiring in the cell, which in turn facilitates tumor development. Here, we review the current knowledge on the interplay between DNA/RNA methylation and metabolic programs in cancer cells. We also discuss the mechanistic role of these pathways in tumor development and progression.
Collapse
Affiliation(s)
- Pallawi Choubey
- Molecular Biology and Genetics Unit (MBGU), Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bangalore, India
| | - Harshdeep Kaur
- Molecular Biology and Genetics Unit (MBGU), Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bangalore, India
| | - Kushagra Bansal
- Molecular Biology and Genetics Unit (MBGU), Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bangalore, India.
| |
Collapse
|
91
|
Smirnova I, Drăghici G, Kazakova O, Vlaia L, Avram S, Mioc A, Mioc M, Macaşoi I, Dehelean C, Voicu A, Şoica C. Hollongdione arylidene derivatives induce antiproliferative activity against melanoma and breast cancer through pro-apoptotic and antiangiogenic mechanisms. Bioorg Chem 2021; 119:105535. [PMID: 34906859 DOI: 10.1016/j.bioorg.2021.105535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 11/27/2021] [Accepted: 11/29/2021] [Indexed: 11/26/2022]
Abstract
The use of natural compounds as starting point for semisynthetic derivatives has already been proven as a valuable source of active anticancer agents. Hollongdione (4,4,8,14-tetramethyl-18-norpregnan-3,20-dion), obtained by few steps from dammarane type triterpenoid dipterocarpol, was chemically modified at C2 and C21 carbon atoms by the Claisen-Schmidt aldol condensation to give a series of arylidene derivatives. The anticancer activity of the obtained compounds was assessed on NCI-60 cancer cell panel, revealing strong antiproliferative effects against a large variety of cancer cells. 2,21-Bis-[3-pyridinyl]-methylidenohollongdione 9 emerged as the most active derivative as indicated by its GI50 values in the micromolar range which, combined with its high selectivity index values, indicated its suitability for deeper biological investigation. The mechanisms involved in compound 9 antiproliferative activity, were investigated through in vitro (DAPI staining) and ex vivo (CAM assay) tests, which exhibited its apoptotic and antiangiogenic activities. In addition, compound 9 showed an overall inhibition of mitochondrial respiration. rtPCR analysis identified the more intimate activity at pro-survival/pro-apoptotic gene level. Collectively, the hollongdione derivative stand as a promising therapeutic option against melanoma and breast cancer provided that future in vivo analysis will certify its clinical efficacy.
Collapse
Affiliation(s)
- Irina Smirnova
- Ufa Institute of Chemistry UFRC RAS, pr. Octyabrya 71, 450054 Ufa, Russian Federation
| | - George Drăghici
- Faculty of Pharmacy, "Victor Babes" University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., Timisoara 300041, Romania; Res. Ctr. Pharmacotoxicol Evaluat, Facculty of Pharmacy, "Victor Babes" University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Sq., Timisoara 300041, Romania
| | - Oxana Kazakova
- Ufa Institute of Chemistry UFRC RAS, pr. Octyabrya 71, 450054 Ufa, Russian Federation.
| | - Lavinia Vlaia
- Faculty of Pharmacy, "Victor Babes" University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., Timisoara 300041, Romania
| | - Stefana Avram
- Faculty of Pharmacy, "Victor Babes" University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., Timisoara 300041, Romania; Res. Ctr. Pharmacotoxicol Evaluat, Facculty of Pharmacy, "Victor Babes" University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Sq., Timisoara 300041, Romania
| | - Alexandra Mioc
- Faculty of Pharmacy, "Victor Babes" University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., Timisoara 300041, Romania; Res. Ctr. Pharmacotoxicol Evaluat, Facculty of Pharmacy, "Victor Babes" University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Sq., Timisoara 300041, Romania
| | - Marius Mioc
- Faculty of Pharmacy, "Victor Babes" University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., Timisoara 300041, Romania; Res. Ctr. Pharmacotoxicol Evaluat, Facculty of Pharmacy, "Victor Babes" University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Sq., Timisoara 300041, Romania
| | - Ioana Macaşoi
- Faculty of Pharmacy, "Victor Babes" University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., Timisoara 300041, Romania; Res. Ctr. Pharmacotoxicol Evaluat, Facculty of Pharmacy, "Victor Babes" University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Sq., Timisoara 300041, Romania
| | - Cristina Dehelean
- Faculty of Pharmacy, "Victor Babes" University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., Timisoara 300041, Romania; Res. Ctr. Pharmacotoxicol Evaluat, Facculty of Pharmacy, "Victor Babes" University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Sq., Timisoara 300041, Romania
| | - Adrian Voicu
- Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., Timisoara 300041, Romania
| | - Codruța Şoica
- Faculty of Pharmacy, "Victor Babes" University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., Timisoara 300041, Romania; Res. Ctr. Pharmacotoxicol Evaluat, Facculty of Pharmacy, "Victor Babes" University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Sq., Timisoara 300041, Romania
| |
Collapse
|
92
|
Cyclodextrin Dispersion of Mebendazole and Flubendazole Improves In Vitro Antiproliferative Activity. Processes (Basel) 2021. [DOI: 10.3390/pr9122185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Mebendazole and flubendazole are antihelmintic drugs that have re-entered the research spotlight due to their exhibited anticancer effects, thus making them strong candidates as repurposed drugs. However, these benzimidazole derivatives exhibit poor solubility in water and various organic solvents, which limits their bioavailability. With the aim of obtaining an improved drug solubility and increased biological effect, mebendazole and flubendazole were complexed with 2-hydroxypropyl-β-cyclodextrin (HPBCD). The binary 1:1 conjugates were physicochemically evaluated by X-ray diffraction, thermal analysis, and FTIR spectroscopy, revealing the formation of physical mixtures. The increased aqueous solubility of the binary 1:1 conjugates vs. pure benzimidazole compounds was demonstrated by performing dissolution tests. The in vitro antiproliferative activity of mebendazole and flubendazole, as well as their combination with HPBCD, was tested on two cancer cell lines, human melanoma—A375 and pulmonary adenocarcinoma—A549 by the MTT assay. The cytotoxic activity manifested in a dose-dependent manner while the presence of HPBCD increased the antiproliferative activity against the targeted cells. Treatment of A375 and A549 cell lines with the binary conjugates induced a significant inhibition of mitochondrial respiration, as revealed by high-resolution respirometry studies. Molecular docking analysis showed that one of the mechanisms related to MEB and FLU cytotoxic activity may be due to the inhibition of MEK/ERK proteins.
Collapse
|
93
|
Duraj T, Carrión-Navarro J, Seyfried TN, García-Romero N, Ayuso-Sacido A. Metabolic therapy and bioenergetic analysis: The missing piece of the puzzle. Mol Metab 2021; 54:101389. [PMID: 34749013 PMCID: PMC8637646 DOI: 10.1016/j.molmet.2021.101389] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/29/2021] [Accepted: 11/01/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Aberrant metabolism is recognized as a hallmark of cancer, a pillar necessary for cellular proliferation. Regarding bioenergetics (ATP generation), most cancers display a preference not only toward aerobic glycolysis ("Warburg effect") and glutaminolysis (mitochondrial substrate level-phosphorylation) but also toward other metabolites such as lactate, pyruvate, and fat-derived sources. These secondary metabolites can assist in proliferation but cannot fully cover ATP demands. SCOPE OF REVIEW The concept of a static metabolic profile is challenged by instances of heterogeneity and flexibility to meet fuel/anaplerotic demands. Although metabolic therapies are a promising tool to improve therapeutic outcomes, either via pharmacological targets or press-pulse interventions, metabolic plasticity is rarely considered. Lack of bioenergetic analysis in vitro and patient-derived models is hindering translational potential. Here, we review the bioenergetics of cancer and propose a simple analysis of major metabolic pathways, encompassing both affordable and advanced techniques. A comprehensive compendium of Seahorse XF bioenergetic measurements is presented for the first time. MAJOR CONCLUSIONS Standardization of principal readouts might help researchers to collect a complete metabolic picture of cancer using the most appropriate methods depending on the sample of interest.
Collapse
Affiliation(s)
- Tomás Duraj
- Faculty of Medicine, Institute for Applied Molecular Medicine (IMMA), CEU San Pablo University, 28668, Madrid, Spain.
| | - Josefa Carrión-Navarro
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223, Madrid, Spain; Brain Tumor Laboratory, Fundación Vithas, Grupo Hospitales Vithas, 28043, Madrid, Spain.
| | - Thomas N Seyfried
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, MA, 02467, USA.
| | - Noemí García-Romero
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223, Madrid, Spain; Brain Tumor Laboratory, Fundación Vithas, Grupo Hospitales Vithas, 28043, Madrid, Spain.
| | - Angel Ayuso-Sacido
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223, Madrid, Spain; Brain Tumor Laboratory, Fundación Vithas, Grupo Hospitales Vithas, 28043, Madrid, Spain; Faculty of Medicine, Universidad Francisco de Vitoria, 28223, Madrid, Spain.
| |
Collapse
|
94
|
Ge Q, Jia D, Cen D, Qi Y, Shi C, Li J, Sang L, Yang LJ, He J, Lin A, Chen S, Wang L. Micropeptide ASAP encoded by LINC00467 promotes colorectal cancer progression by directly modulating ATP synthase activity. J Clin Invest 2021; 131:152911. [PMID: 34591791 DOI: 10.1172/jci152911] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 09/23/2021] [Indexed: 02/06/2023] Open
Abstract
Emerging evidence has shown that open reading frames inside long noncoding RNAs (lncRNAs) could encode micropeptides. However, their roles in cellular energy metabolism and tumor progression remain largely unknown. Here, we identified a 94 amino acid-length micropeptide encoded by lncRNA LINC00467 in colorectal cancer. We also characterized its conservation across higher mammals, localization to mitochondria, and the concerted local functions. This peptide enhanced the ATP synthase construction by interacting with the subunits α and γ (ATP5A and ATP5C), increased ATP synthase activity and mitochondrial oxygen consumption rate, and thereby promoted colorectal cancer cell proliferation. Hence, this micropeptide was termed ATP synthase-associated peptide (ASAP). Furthermore, loss of ASAP suppressed patient-derived xenograft growth with attenuated ATP synthase activity and mitochondrial ATP production. Clinically, high expression of ASAP and LINC00467 predicted poor prognosis of colorectal cancer patients. Taken together, our findings revealed a colorectal cancer-associated micropeptide as a vital player in mitochondrial metabolism and provided a therapeutic target for colorectal cancer.
Collapse
Affiliation(s)
- Qiwei Ge
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China.,Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province, China.,MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Dingjiacheng Jia
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China.,Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Dong Cen
- Department of General Surgery and
| | - Yadong Qi
- Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province, China.,Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Chengyu Shi
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang Province, China.,Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang, China
| | - Junhong Li
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang Province, China.,Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang, China
| | - Lingjie Sang
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang Province, China.,Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang, China
| | - Luo-Jia Yang
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang Province, China.,Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang, China
| | - Jiamin He
- Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province, China.,Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Aifu Lin
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang Province, China.,Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang, China.,Cancer Center, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Shujie Chen
- Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province, China.,Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang Province, China.,Cancer Center, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Liangjing Wang
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China.,Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province, China.,Cancer Center, Zhejiang University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
95
|
Foo BJA, Eu JQ, Hirpara JL, Pervaiz S. Interplay between Mitochondrial Metabolism and Cellular Redox State Dictates Cancer Cell Survival. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:1341604. [PMID: 34777681 PMCID: PMC8580634 DOI: 10.1155/2021/1341604] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 09/30/2021] [Accepted: 10/04/2021] [Indexed: 02/06/2023]
Abstract
Mitochondria are the main powerhouse of the cell, generating ATP through the tricarboxylic acid cycle (TCA) and oxidative phosphorylation (OXPHOS), which drives myriad cellular processes. In addition to their role in maintaining bioenergetic homeostasis, changes in mitochondrial metabolism, permeability, and morphology are critical in cell fate decisions and determination. Notably, mitochondrial respiration coupled with the passage of electrons through the electron transport chain (ETC) set up a potential source of reactive oxygen species (ROS). While low to moderate increase in intracellular ROS serves as secondary messenger, an overwhelming increase as a result of either increased production and/or deficient antioxidant defenses is detrimental to biomolecules, cells, and tissues. Since ROS and mitochondria both regulate cell fate, attention has been drawn to their involvement in the various processes of carcinogenesis. To that end, the link between a prooxidant milieu and cell survival and proliferation as well as a switch to mitochondrial OXPHOS associated with recalcitrant cancers provide testimony for the remarkable metabolic plasticity as an important hallmark of cancers. In this review, the regulation of cell redox status by mitochondrial metabolism and its implications for cancer cell fate will be discussed followed by the significance of mitochondria-targeted therapies for cancer.
Collapse
Affiliation(s)
- Brittney Joy-Anne Foo
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, Singapore
| | - Jie Qing Eu
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, Singapore
- Cancer Science Institute, NUS, Singapore, Singapore
| | | | - Shazib Pervaiz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, NUS, Singapore, Singapore
- NUS Medicine Healthy Longevity Program, Yong Loo Lin School of Medicine, NUS, Singapore, Singapore
- Integrative Sciences and Engineering Program, NUS Graduate School, NUS, Singapore, Singapore
- National University Cancer Institute, National University Health System, Singapore, Singapore
- Faculté de Médicine, Université de Paris, Paris, France
| |
Collapse
|
96
|
Iorio M, Umesh Ganesh N, De Luise M, Porcelli AM, Gasparre G, Kurelac I. The Neglected Liaison: Targeting Cancer Cell Metabolic Reprogramming Modifies the Composition of Non-Malignant Populations of the Tumor Microenvironment. Cancers (Basel) 2021; 13:cancers13215447. [PMID: 34771610 PMCID: PMC8582418 DOI: 10.3390/cancers13215447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/27/2021] [Accepted: 10/28/2021] [Indexed: 11/16/2022] Open
Abstract
Metabolic reprogramming is a well-known hallmark of cancer, whereby the development of drugs that target cancer cell metabolism is gaining momentum. However, when establishing preclinical studies and clinical trials, it is often neglected that a tumor mass is a complex system in which cancer cells coexist and interact with several types of microenvironment populations, including endothelial cells, fibroblasts and immune cells. We are just starting to understand how such populations are affected by the metabolic changes occurring in a transformed cell and little is known about the impact of metabolism-targeting drugs on the non-malignant tumor components. Here we provide a general overview of the links between cancer cell metabolism and tumor microenvironment (TME), particularly focusing on the emerging literature reporting TME-specific effects of metabolic therapies.
Collapse
Affiliation(s)
- Maria Iorio
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (M.I.); (N.U.G.); (M.D.L.); (G.G.)
- Center for Applied Biomedical Research, University of Bologna, 40138 Bologna, Italy;
- Centro Studi e Ricerca sulle Neoplasie Ginecologiche (CSR), University of Bologna, 40138 Bologna, Italy
| | - Nikkitha Umesh Ganesh
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (M.I.); (N.U.G.); (M.D.L.); (G.G.)
- Center for Applied Biomedical Research, University of Bologna, 40138 Bologna, Italy;
- Centro Studi e Ricerca sulle Neoplasie Ginecologiche (CSR), University of Bologna, 40138 Bologna, Italy
| | - Monica De Luise
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (M.I.); (N.U.G.); (M.D.L.); (G.G.)
- Center for Applied Biomedical Research, University of Bologna, 40138 Bologna, Italy;
- Centro Studi e Ricerca sulle Neoplasie Ginecologiche (CSR), University of Bologna, 40138 Bologna, Italy
| | - Anna Maria Porcelli
- Center for Applied Biomedical Research, University of Bologna, 40138 Bologna, Italy;
- Centro Studi e Ricerca sulle Neoplasie Ginecologiche (CSR), University of Bologna, 40138 Bologna, Italy
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, 40126 Bologna, Italy
- Interdepartmental Center of Industrial Research (CIRI) Life Science and Health Technologies, University of Bologna, 40064 Ozzano dell’Emilia, Italy
| | - Giuseppe Gasparre
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (M.I.); (N.U.G.); (M.D.L.); (G.G.)
- Center for Applied Biomedical Research, University of Bologna, 40138 Bologna, Italy;
- Centro Studi e Ricerca sulle Neoplasie Ginecologiche (CSR), University of Bologna, 40138 Bologna, Italy
| | - Ivana Kurelac
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (M.I.); (N.U.G.); (M.D.L.); (G.G.)
- Center for Applied Biomedical Research, University of Bologna, 40138 Bologna, Italy;
- Centro Studi e Ricerca sulle Neoplasie Ginecologiche (CSR), University of Bologna, 40138 Bologna, Italy
- Correspondence: ; Tel.: +39-051-2088-418
| |
Collapse
|
97
|
Xie Y, Zhang W, Guo L, Kril LM, Begley KL, Sviripa VM, Chen X, Liu X, Lee EY, He D, Wang C, Gao T, Liu X, Evers BM, Watt DS, Liu C. Potent Synergistic Effect on C-Myc-Driven Colorectal Cancers Using a Novel Indole-Substituted Quinoline with a Plk1 Inhibitor. Mol Cancer Ther 2021; 20:1893-1903. [PMID: 34376582 PMCID: PMC8492540 DOI: 10.1158/1535-7163.mct-20-1017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 03/24/2021] [Accepted: 07/23/2021] [Indexed: 11/16/2022]
Abstract
Developing effective treatments for colorectal cancers through combinations of small-molecule approaches and immunotherapies present intriguing possibilities for managing these otherwise intractable cancers. During a broad-based, screening effort against multiple colorectal cancer cell lines, we identified indole-substituted quinolines (ISQ), such as N7,N7 -dimethyl-3-(1-methyl-1H-indol-3-yl)quinoline-2,7-diamine (ISQ-1), as potent in vitro inhibitors of several cancer cell lines. We found that ISQ-1 inhibited Wnt signaling, a main driver in the pathway governing colorectal cancer development, and ISQ-1 also activated adenosine monophosphate kinase (AMPK), a cellular energy-homeostasis master regulator. We explored the effect of ISQs on cell metabolism. Seahorse assays measuring oxygen consumption rate (OCR) indicated that ISQ-1 inhibited complex I (i.e., NADH ubiquinone oxidoreductase) in the mitochondrial, electron transport chain (ETC). In addition, ISQ-1 treatment showed remarkable synergistic depletion of oncogenic c-Myc protein level in vitro and induced strong tumor remission in vivo when administered together with BI2536, a polo-like kinase-1 (Plk1) inhibitor. These studies point toward the potential value of dual drug therapies targeting the ETC and Plk-1 for the treatment of c-Myc-driven cancers.
Collapse
Affiliation(s)
- Yanqi Xie
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, Kentucky
- Lucille Parker Markey Cancer Center, University of Kentucky, Lexington, Kentucky
| | - Wen Zhang
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, Kentucky
- Lucille Parker Markey Cancer Center, University of Kentucky, Lexington, Kentucky
| | - Lichao Guo
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, Kentucky
- Lucille Parker Markey Cancer Center, University of Kentucky, Lexington, Kentucky
- Center for Drug Innovation and Discovery, Hebei Normal University, Shijiazhuang, Hebei, People's Republic of China
| | - Liliia M Kril
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, Kentucky
- Lucille Parker Markey Cancer Center, University of Kentucky, Lexington, Kentucky
- Center for Pharmaceutical Research and Innovation, College of Pharmacy, University of Kentucky, Lexington, Kentucky
| | - Kristin L Begley
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, Kentucky
- Lucille Parker Markey Cancer Center, University of Kentucky, Lexington, Kentucky
- Center for Pharmaceutical Research and Innovation, College of Pharmacy, University of Kentucky, Lexington, Kentucky
| | - Vitaliy M Sviripa
- Center for Drug Innovation and Discovery, Hebei Normal University, Shijiazhuang, Hebei, People's Republic of China
- Center for Pharmaceutical Research and Innovation, College of Pharmacy, University of Kentucky, Lexington, Kentucky
| | - Xi Chen
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, Kentucky
- Lucille Parker Markey Cancer Center, University of Kentucky, Lexington, Kentucky
- Center for Drug Innovation and Discovery, Hebei Normal University, Shijiazhuang, Hebei, People's Republic of China
| | - Xifu Liu
- Center for Drug Innovation and Discovery, Hebei Normal University, Shijiazhuang, Hebei, People's Republic of China
| | - Eun Y Lee
- Lucille Parker Markey Cancer Center, University of Kentucky, Lexington, Kentucky
- Department of Pathology & Laboratory Medicine, College of Medicine, University of Kentucky, Lexington, Kentucky
| | - Daheng He
- Lucille Parker Markey Cancer Center, University of Kentucky, Lexington, Kentucky
| | - Chi Wang
- Lucille Parker Markey Cancer Center, University of Kentucky, Lexington, Kentucky
| | - Tianyan Gao
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, Kentucky
- Lucille Parker Markey Cancer Center, University of Kentucky, Lexington, Kentucky
| | - Xiaoqi Liu
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, Kentucky
| | - B Mark Evers
- Lucille Parker Markey Cancer Center, University of Kentucky, Lexington, Kentucky
- Department of Surgery, College of Medicine, University of Kentucky, Lexington, Kentucky
| | - David S Watt
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, Kentucky.
- Lucille Parker Markey Cancer Center, University of Kentucky, Lexington, Kentucky
- Center for Pharmaceutical Research and Innovation, College of Pharmacy, University of Kentucky, Lexington, Kentucky
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky
| | - Chunming Liu
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, Kentucky.
- Lucille Parker Markey Cancer Center, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
98
|
Identification of Possible Salivary Metabolic Biomarkers and Altered Metabolic Pathways in South American Patients Diagnosed with Oral Squamous Cell Carcinoma. Metabolites 2021; 11:metabo11100650. [PMID: 34677365 PMCID: PMC8537096 DOI: 10.3390/metabo11100650] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/10/2021] [Accepted: 08/10/2021] [Indexed: 12/14/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) represents 90% of oral malignant neoplasms. The search for specific biomarkers for OSCC is a very active field of research contributing to establishing early diagnostic methods and unraveling underlying pathogenic mechanisms. In this work we investigated the salivary metabolites and the metabolic pathways of OSCC aiming find possible biomarkers. Salivary metabolites samples from 27 OSCC patients and 41 control individuals were compared through a gas chromatography coupled to a mass spectrometer (GC-MS) technique. Our results allowed identification of pathways of the malate-aspartate shuttle, the beta-alanine metabolism, and the Warburg effect. The possible salivary biomarkers were identified using the area under receiver-operating curve (AUC) criterion. Twenty-four metabolites were identified with AUC > 0.8. Using the threshold of AUC = 0.9 we find malic acid, maltose, protocatechuic acid, lactose, 2-ketoadipic, and catechol metabolites expressed. We notice that this is the first report of salivary metabolome in South American oral cancer patients, to the best of our knowledge. Our findings regarding these metabolic changes are important in discovering salivary biomarkers of OSCC patients. However, additional work needs to be performed considering larger populations to validate our results.
Collapse
|
99
|
Jariwala N, Mehta GA, Bhatt V, Hussein S, Parker KA, Yunus N, Parker JS, Guo JY, Gatza ML. CPT1A and fatty acid β-oxidation are essential for tumor cell growth and survival in hormone receptor-positive breast cancer. NAR Cancer 2021; 3:zcab035. [PMID: 34514415 PMCID: PMC8428294 DOI: 10.1093/narcan/zcab035] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 08/18/2021] [Accepted: 08/25/2021] [Indexed: 12/19/2022] Open
Abstract
Chromosome 11q13-14 amplification is a defining feature of high-risk hormone receptor-positive (HR+) breast cancer; however, the mechanism(s) by which this amplicon contributes to breast tumorigenesis remains unclear. In the current study, proteogenomic analyses of >3000 breast tumors from the TCGA, METABRIC and CPTAC studies demonstrated that carnitine palmitoyltransferase 1A (CPT1A), which is localized to this amplicon, is overexpressed at the mRNA and protein level in aggressive luminal tumors, strongly associated with indicators of tumor proliferation and a predictor of poor prognosis. In vitro genetic studies demonstrated that CPT1A is required for and can promote luminal breast cancer proliferation, survival, as well as colony and mammosphere formation. Since CPT1A is the rate-limiting enzyme during fatty acid oxidation (FAO), our data indicate that FAO may be essential for these tumors. Pharmacologic inhibition of FAO prevented in vitro and in vivo tumor growth and cell proliferation as well as promoted apoptosis in luminal breast cancer cells and orthotopic xenograft tumor models. Collectively, our data establish an oncogenic role for CPT1A and FAO in HR+ luminal tumors and provide preclinical evidence and rationale supporting further investigation of FAO as a potential therapeutic opportunity for the treatment of HR+ breast cancer.
Collapse
Affiliation(s)
- Nidhi Jariwala
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08903, USA
| | - Gaurav A Mehta
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08903, USA
| | - Vrushank Bhatt
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08903, USA
| | - Shaimaa Hussein
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08903, USA
| | - Kimberly A Parker
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08903, USA
| | - Neha Yunus
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08903, USA
| | - Joel S Parker
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill NC, 27599, USA
| | | | - Michael L Gatza
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08903, USA
| |
Collapse
|
100
|
Bedaquiline, an FDA-approved drug, inhibits mitochondrial ATP production and metastasis in vivo, by targeting the gamma subunit (ATP5F1C) of the ATP synthase. Cell Death Differ 2021; 28:2797-2817. [PMID: 33986463 PMCID: PMC8408289 DOI: 10.1038/s41418-021-00788-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 04/09/2021] [Accepted: 04/15/2021] [Indexed: 02/08/2023] Open
Abstract
Here, we provide evidence that high ATP production by the mitochondrial ATP-synthase is a new therapeutic target for anticancer therapy, especially for preventing tumor progression. More specifically, we isolated a subpopulation of ATP-high cancer cells which are phenotypically aggressive and demonstrate increases in proliferation, stemness, anchorage-independence, cell migration, invasion and multi-drug resistance, as well as high antioxidant capacity. Clinically, these findings have important implications for understanding treatment failure and cancer cell dormancy. Using bioinformatic analysis of patient samples, we defined a mitochondrial-related gene signature for metastasis, which features the gamma-subunit of the mitochondrial ATP-synthase (ATP5F1C). The relationship between ATP5F1C protein expression and metastasis was indeed confirmed by immunohistochemistry. Next, we used MDA-MB-231 cells as a model system to functionally validate these findings. Importantly, ATP-high MDA-MB-231 cells showed a nearly fivefold increase in metastatic capacity in vivo. Consistent with these observations, ATP-high cells overexpressed (i) components of mitochondrial complexes I-V, including ATP5F1C, and (ii) markers associated with circulating tumor cells (CTCs) and metastasis, such as EpCAM and VCAM1. Knockdown of ATP5F1C expression significantly reduced ATP-production, anchorage-independent growth, and cell migration, as predicted. Similarly, therapeutic administration of the FDA-approved drug, Bedaquiline, downregulated ATP5F1C expression in vitro and prevented spontaneous metastasis in vivo. In contrast, Bedaquiline had no effect on the growth of non-tumorigenic mammary epithelial cells (MCF10A) or primary tumors in vivo. Taken together, our results suggest that mitochondrial ATP depletion is a new therapeutic strategy for metastasis prophylaxis, to avoid treatment failure. In summary, we conclude that mitochondrial ATP5F1C is a promising new biomarker and molecular target for future drug development, for the prevention of metastatic disease progression.
Collapse
|