1
|
Crossley SW, Tenney L, Pham VN, Xie X, Zhao MW, Chang CJ. A Transfer Hydrogenation Approach to Activity-Based Sensing of Formate in Living Cells. J Am Chem Soc 2024; 146:8865-8876. [PMID: 38470125 PMCID: PMC11487638 DOI: 10.1021/jacs.3c09735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Formate is a major reactive carbon species in one-carbon metabolism, where it serves as an endogenous precursor for amino acid and nucleic acid biosynthesis and a cellular source of NAD(P)H. On the other hand, aberrant elevations in cellular formate are connected to progression of serious diseases, including cancer and Alzheimer's disease. Traditional methods for formate detection in biological environments often rely on sample destruction or extensive processing, resulting in a loss of spatiotemporal information. To help address these limitations, here we present the design, synthesis, and biological evaluation of a first-generation activity-based sensing system for live-cell formate imaging that relies on iridium-mediated transfer hydrogenation chemistry. Formate facilitates an aldehyde-to-alcohol conversion on various fluorophore scaffolds to enable fluorescence detection of this one-carbon unit, including through a two-color ratiometric response with internal calibration. The resulting two-component probe system can detect changes in formate levels in living cells with a high selectivity over potentially competing biological analytes. Moreover, this activity-based sensing system can visualize changes in endogenous formate fluxes through alterations of one-carbon pathways in cell-based models of human colon cancer, presaging the potential utility of this chemical approach to probe the continuum between one-carbon metabolism and signaling in cancer and other diseases.
Collapse
Affiliation(s)
- Steven W.M. Crossley
- Department of Chemistry, University of California, Berkeley, Berkeley, California, 94720, United States
| | - Logan Tenney
- Department of Chemistry, University of California, Berkeley, Berkeley, California, 94720, United States
| | - Vanha N. Pham
- Department of Chemistry, University of California, Berkeley, Berkeley, California, 94720, United States
| | - Xiao Xie
- Department of Chemistry, University of California, Berkeley, Berkeley, California, 94720, United States
| | - Michelle W. Zhao
- Department of Chemistry, University of California, Berkeley, Berkeley, California, 94720, United States
| | - Christopher J. Chang
- Department of Chemistry, University of California, Berkeley, Berkeley, California, 94720, United States
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California, 94720, United States
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California, 94720, United States
| |
Collapse
|
2
|
Srivastava J, Trivedi R, Saxena P, Yadav S, Gupta R, Nityanand S, Kumar D, Chaturvedi CP. Bone marrow plasma metabonomics of idiopathic acquired aplastic anemia patients using 1H nuclear magnetic resonance spectroscopy. Metabolomics 2023; 19:94. [PMID: 37975930 DOI: 10.1007/s11306-023-02056-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 10/10/2023] [Indexed: 11/19/2023]
Abstract
INTRODUCTION Idiopathic acquired aplastic anemia (AA) is a bone marrow failure disorder where aberrant T-cell functions lead to depletion of hematopoietic stem and progenitor cells in the bone marrow (BM) microenvironment. T-cells undergo metabolic rewiring, which regulates their proliferation and differentiation. Therefore, studying metabolic variation in AA patients may aid us with a better understanding of the T-cell regulatory pathways governed by metabolites and their pathological engagement in the disease. OBJECTIVE To identify the differential metabolites in BM plasma of AA patients, AA follow-up (AAF) in comparison to normal controls (NC) and to identify potential disease biomarker(s). METHODS The study used 1D 1H NMR Carr-Purcell-Meiboom-Gill (CPMG) spectra to identify the metabolites present in the BM plasma samples of AA (n = 40), AAF (n = 16), and NC (n = 20). Metabolic differences between the groups and predictive biomarkers were identified by using multivariate analysis and receiver operating characteristic (ROC) module of Metaboanalyst V5.0 tool, respectively. RESULTS The AA and AAF samples were well discriminated from NC group as per Principal Component analysis (PCA). Further, we found significant alteration in the levels of 17 metabolites in AA involved in amino-acid (Leucine, serine, threonine, phenylalanine, lysine, histidine, valine, tyrosine, and proline), carbohydrate (Glucose, lactate and mannose), fatty acid (Acetate, glycerol myo-inositol and citrate), and purine metabolism (hypoxanthine) in comparison to NC. Additionally, biomarker analysis predicted Hypoxanthine and Acetate can be used as a potential biomarker. CONCLUSION The study highlights the significant metabolic alterations in the BM plasma of AA patients which may have implication in the disease pathobiology.
Collapse
Affiliation(s)
- Jyotika Srivastava
- Department of Hematology, Stem Cell Research Centre, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Rae Barely Road, Lucknow, Uttar Pradesh, 226014, India
| | - Rimjhim Trivedi
- Centre of Biomedical Research (CBMR), Sanjay Gandhi Post Graduate Institute of Medical Sciences Campus, Rae Barely Road, Lucknow, Uttar Pradesh, 226014, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Pragati Saxena
- Department of Hematology, Stem Cell Research Centre, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Rae Barely Road, Lucknow, Uttar Pradesh, 226014, India
| | - Sanjeev Yadav
- Department of Hematology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Rae Barely Road, Lucknow, Uttar Pradesh, 226014, India
| | - Ruchi Gupta
- Department of Hematology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Rae Barely Road, Lucknow, Uttar Pradesh, 226014, India
| | - Soniya Nityanand
- Department of Hematology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Rae Barely Road, Lucknow, Uttar Pradesh, 226014, India
| | - Dinesh Kumar
- Centre of Biomedical Research (CBMR), Sanjay Gandhi Post Graduate Institute of Medical Sciences Campus, Rae Barely Road, Lucknow, Uttar Pradesh, 226014, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India.
| | - Chandra P Chaturvedi
- Department of Hematology, Stem Cell Research Centre, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Rae Barely Road, Lucknow, Uttar Pradesh, 226014, India.
| |
Collapse
|
3
|
Delbrouck C, Kiweler N, Chen O, Pozdeev VI, Haase L, Neises L, Oudin A, Fouquier d'Hérouël A, Shen R, Schlicker L, Halder R, Lesur A, Schuster A, Lorenz NI, Jaeger C, Feucherolles M, Frache G, Szpakowska M, Chevigne A, Ronellenfitsch MW, Moussay E, Piraud M, Skupin A, Schulze A, Niclou SP, Letellier E, Meiser J. Formate promotes invasion and metastasis in reliance on lipid metabolism. Cell Rep 2023; 42:113034. [PMID: 37651228 DOI: 10.1016/j.celrep.2023.113034] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/09/2023] [Accepted: 08/11/2023] [Indexed: 09/02/2023] Open
Abstract
Metabolic rewiring is essential for cancer onset and progression. We previously showed that one-carbon metabolism-dependent formate production often exceeds the anabolic demand of cancer cells, resulting in formate overflow. Furthermore, we showed that increased extracellular formate concentrations promote the in vitro invasiveness of glioblastoma cells. Here, we substantiate these initial observations with ex vivo and in vivo experiments. We also show that exposure to exogeneous formate can prime cancer cells toward a pro-invasive phenotype leading to increased metastasis formation in vivo. Our results suggest that the increased local formate concentration within the tumor microenvironment can be one factor to promote metastases. Additionally, we describe a mechanistic interplay between formate-dependent increased invasiveness and adaptations of lipid metabolism and matrix metalloproteinase activity. Our findings consolidate the role of formate as pro-invasive metabolite and warrant further research to better understand the interplay between formate and lipid metabolism.
Collapse
Affiliation(s)
- Catherine Delbrouck
- Cancer Metabolism Group, Department of Cancer Research, Luxembourg Institute of Health, 1210 Luxembourg, Luxembourg; Faculty of Science, Technology and Medicine, University of Luxembourg, 2 avenue de Université, 4362 Esch-sur-Alzette, Luxembourg
| | - Nicole Kiweler
- Cancer Metabolism Group, Department of Cancer Research, Luxembourg Institute of Health, 1210 Luxembourg, Luxembourg
| | - Oleg Chen
- Cancer Metabolism Group, Department of Cancer Research, Luxembourg Institute of Health, 1210 Luxembourg, Luxembourg
| | - Vitaly I Pozdeev
- Molecular Disease Mechanisms Group, Faculty of Science, Technology and Medicine, University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg
| | - Lara Haase
- Cancer Metabolism Group, Department of Cancer Research, Luxembourg Institute of Health, 1210 Luxembourg, Luxembourg; Faculty of Science, Technology and Medicine, University of Luxembourg, 2 avenue de Université, 4362 Esch-sur-Alzette, Luxembourg
| | - Laura Neises
- Cancer Metabolism Group, Department of Cancer Research, Luxembourg Institute of Health, 1210 Luxembourg, Luxembourg
| | - Anaïs Oudin
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health, 1210 Luxembourg, Luxembourg
| | - Aymeric Fouquier d'Hérouël
- Integrative Cell Signaling Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg
| | - Ruolin Shen
- Helmholtz AI Central Unit, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Lisa Schlicker
- Division of Tumor Metabolism and Microenvironment, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Proteomics Core Facility, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Rashi Halder
- RNAseq Platform, Systems Ecology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg
| | - Antoine Lesur
- Cancer Metabolism Group, Department of Cancer Research, Luxembourg Institute of Health, 1210 Luxembourg, Luxembourg
| | - Anne Schuster
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health, 1210 Luxembourg, Luxembourg
| | - Nadja I Lorenz
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany; German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany; Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, 60596 Frankfurt am Main, Germany
| | - Christian Jaeger
- Metabolomics Platform, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg
| | - Maureen Feucherolles
- Molecular and Thermal Analysis Group, Materials Research and Technology, Luxembourg Institute of Science and Technology, 4422 Belvaux, Luxembourg
| | - Gilles Frache
- Molecular and Thermal Analysis Group, Materials Research and Technology, Luxembourg Institute of Science and Technology, 4422 Belvaux, Luxembourg
| | - Martyna Szpakowska
- Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg
| | - Andy Chevigne
- Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg
| | - Michael W Ronellenfitsch
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany; German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany; Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, 60596 Frankfurt am Main, Germany; University Cancer Center Frankfurt (UCT), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany
| | - Etienne Moussay
- Tumor-Stroma Interactions Group, Department of Cancer Research, Luxembourg Institute of Health, 1210 Luxembourg, Luxembourg
| | - Marie Piraud
- Helmholtz AI Central Unit, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Alexander Skupin
- Integrative Cell Signaling Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg; Department of Neurosciences, University of California San Diego, La Jolla, CA 92092, USA; Department of Physics and Material Science, University of Luxembourg, 1511 Luxembourg, Luxembourg
| | - Almut Schulze
- Division of Tumor Metabolism and Microenvironment, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Simone P Niclou
- Faculty of Science, Technology and Medicine, University of Luxembourg, 2 avenue de Université, 4362 Esch-sur-Alzette, Luxembourg; NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health, 1210 Luxembourg, Luxembourg
| | - Elisabeth Letellier
- Molecular Disease Mechanisms Group, Faculty of Science, Technology and Medicine, University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg
| | - Johannes Meiser
- Cancer Metabolism Group, Department of Cancer Research, Luxembourg Institute of Health, 1210 Luxembourg, Luxembourg.
| |
Collapse
|
4
|
Ferdous MRU, Abdalla M, Yang M, Xiaoling L, Song Y. Berberine chloride (dual topoisomerase I and II inhibitor) modulate mitochondrial uncoupling protein (UCP1) in molecular docking and dynamic with in-vitro cytotoxic and mitochondrial ATP production. J Biomol Struct Dyn 2023; 41:1704-1714. [PMID: 35612892 DOI: 10.1080/07391102.2021.2024255] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Obesity initiates numerous diseases like cardiovascular, metabolic, and type 2 diabetes, and obesity is a vital cause of death worldwide. Plants are necessary to the source of life. Several drug compounds isolated from plants are called phytochemicals which are safe, effective drug moieties to treat several diseases. Berberine chloride is a dual topoisomerase I and II inhibitor, that exhibited potent antitumor activities against several malignancies. However, the effect of Berberine on mitochondria remains unknown. The focus of this study was to determine the role of Berberine on mitochondrial uncoupling protein (UCP1), ATP production, and cytotoxic effect of HEK293T cell at a time and dose-dependent manner analysis by CCK8 assay. The upregulation of mitochondrial UCP1 gene expression reduces adipocyte content by initiating thermogenesis. In this study, berberine chloride significantly up-regulates UCP1 gene expression in brown adipocytes. AT 10 µM concentration of Berberine 48 h treatment demonstrated significant cell death. The decreased level of ATP production leads to mitochondrial uncoupling. Initiate thermogenesis reducing fat droplets in adipocytes. The first time, we used molecular docking and dynamic of Berberine with UCP1 gene in this study and revealed therapeutic potential of Berberine via modulation of mitochondrial UCP1 gene. Further investigation will reveal new insight into mechanisms to treat metabolic-related diseases.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Md Reyad-Ul Ferdous
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.,Department of Internal Medicine, Cheeloo College of Medicine Shandong University, Jinan, Shandong, China.,Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, China.,Shandong Institute of Endocrinology & Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong, China
| | - Mohnad Abdalla
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, PR China
| | - Mengjiao Yang
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital, Shandong First Medical University, Shandong, China
| | - Li Xiaoling
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital, Shandong First Medical University, Shandong, China
| | - Yongfeng Song
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.,Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, China.,Shandong Institute of Endocrinology & Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong, China.,Department of Endocrinology and Metabolism, Shandong Provincial Hospital, Shandong First Medical University, Shandong, China
| |
Collapse
|
5
|
Zhu W, Wang C, Xue L, Liu L, Yang X, Liu Z, Zhang S, Luo D. The SMYD3-MTHFD1L-formate metabolic regulatory axis mediates mitophagy to inhibit M1 polarization in macrophages. Int Immunopharmacol 2022; 113:109352. [DOI: 10.1016/j.intimp.2022.109352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 10/07/2022] [Accepted: 10/11/2022] [Indexed: 11/05/2022]
|
6
|
Okoye CN, Chinnappareddy N, Stevens D, Kamunde C. Factors affecting liver mitochondrial hydrogen peroxide emission. Comp Biochem Physiol B Biochem Mol Biol 2022; 259:110713. [PMID: 35026417 DOI: 10.1016/j.cbpb.2022.110713] [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: 06/15/2021] [Revised: 12/19/2021] [Accepted: 01/04/2022] [Indexed: 10/19/2022]
Abstract
Mitochondria are key cellular sources of reactive oxygen species (ROS) and contain at least 12 known sites on multiple enzymes that convert molecular oxygen to superoxide and hydrogen peroxide (H2O2). Quantitation of site-specific ROS emission is critical to understand the relative contribution of different sites and the pathophysiologic importance of mitochondrial ROS. However, factors that affect mitochondrial ROS emission are not well understood. We characterized and optimized conditions for maximal total and site-specific H2O2 emission during oxidation of standard substrates and probed the source of the high H2O2 emission in unenergized rainbow trout liver mitochondria. We found that mitochondrial H2O2 emission capacity depended on the substrate being oxidized, mitochondrial protein concentration, and composition of the ROS detection system. Contrary to our expectation, addition of exogenous superoxide dismutase reduced H2O2 emission. Titration of conventional mitochondrial electron transfer system (ETS) inhibitors over a range of conditions revealed that one size does not fit all; inhibitor concentrations evoking maximal responses varied with substrate and were moderated by the presence of other inhibitors. Moreover, the efficacy of suppressors of electron leak (S1QEL1.1 and S3QEL2) was low and depended on the substrate being oxidized. We found that H2O2 emission in unenergized rainbow trout liver mitochondria was suppressed by GKT136901 suggesting that it is associated with NADPH oxidase activity. We conclude that optimization of assay conditions is critical for quantitation of maximal H2O2 emission and would facilitate more valid comparisons of mitochondrial total and site-specific H2O2 emission capacities between studies, tissues, and species.
Collapse
Affiliation(s)
- Chidozie N Okoye
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, PE C1A 4P3, Canada
| | - Nirmala Chinnappareddy
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, PE C1A 4P3, Canada
| | - Don Stevens
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, PE C1A 4P3, Canada
| | - Collins Kamunde
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, PE C1A 4P3, Canada.
| |
Collapse
|
7
|
Udumula MP, Sakr S, Dar S, Alvero AB, Ali-Fehmi R, Abdulfatah E, Li J, Jiang J, Tang A, Buekers T, Morris R, Munkarah A, Giri S, Rattan R. Ovarian cancer modulates the immunosuppressive function of CD11b +Gr1 + myeloid cells via glutamine metabolism. Mol Metab 2021; 53:101272. [PMID: 34144215 PMCID: PMC8267600 DOI: 10.1016/j.molmet.2021.101272] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/03/2021] [Accepted: 06/11/2021] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE Immature CD11b + Gr1+ myeloid cells that acquire immunosuppressive capability, also known as myeloid-derived suppressor cells (MDSCs), are a heterogeneous population of cells that regulate immune responses. Our study's objective was to elucidate the role of ovarian cancer microenvironment in regulating the immunosuppressive function of CD11b+Gr1+ myeloid cells. METHODS All studies were performed using the intraperitoneal ID8 syngeneic epithelial ovarian cancer mouse model. Myeloid cell depletion and immunotherapy were carried out using anti-Gr1 mAb, gemcitabine treatments, and/or anti-PD1 mAb. The treatment effect was assessed by a survival curve, in situ luciferase-guided imaging, and histopathologic evaluation. Adoptive transfer assays were carried out between congenic CD45.2 and CD45.1 mice. Immune surface and intracellular markers were assessed by flow cytometry. ELISA, western blot, and RT-PCR techniques were employed to assess the protein and RNA expression of various markers. Bone marrow-derived myeloid cells were used for ex-vivo studies. RESULTS The depletion of Gr1+ immunosuppressive myeloid cells alone and in combination with anti-PD1 immunotherapy inhibited ovarian cancer growth. In addition to the adoptive transfer studies, these findings validate the role of immunosuppressive CD11b+Gr1+ myeloid cells in promoting ovarian cancer. Mechanistic investigations showed that ID8 tumor cells and their microenvironments produced recruitment and regulatory factors for immunosuppressive CD11b+Gr1+ myeloid cells. CD11b+Gr1+ myeloid cells primed by ID8 tumors showed increased immunosuppressive marker expression and acquired an energetic metabolic phenotype promoted primarily by increased oxidative phosphorylation fueled by glutamine. Inhibiting the glutamine metabolic pathway reduced the increased oxidative phosphorylation and decreased immunosuppressive markers' expression and function. Dihydrolipoamide succinyl transferase (DLST), a subunit of α-KGDC in the TCA cycle, was found to be the most significantly elevated gene in tumor-primed myeloid cells. The inhibition of DLST reduced oxidative phosphorylation, immunosuppressive marker expression and function in myeloid cells. CONCLUSION Our study shows that the ovarian cancer microenvironment can regulate the metabolism and function of immunosuppressive CD11b + Gr1+ myeloid cells and modulate its immune microenvironment. Targeting glutamine metabolism via DLST in immunosuppressive myeloid cells decreased their activity, leading to a reduction in the immunosuppressive tumor microenvironment. Thus, targeting glutamine metabolism has the potential to enhance the success of immunotherapy in ovarian cancer.
Collapse
Affiliation(s)
- Mary P Udumula
- Division of Gynecology Oncology, Department of Women's Health Services, Henry Ford Cancer Institute and Henry Ford Health System, Detroit, MI, USA
| | - Sharif Sakr
- Department of Gynecology Oncology, Barbara Ann Karmanos Cancer Institute and Wayne State University, Detroit, MI, USA
| | - Sajad Dar
- Division of Gynecology Oncology, Department of Women's Health Services, Henry Ford Cancer Institute and Henry Ford Health System, Detroit, MI, USA
| | - Ayesha B Alvero
- Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| | - Rouba Ali-Fehmi
- Department of Pathology, Wayne State University and Barbara Ann Karmanos Cancer Institute, Detroit, MI, USA
| | - Eman Abdulfatah
- Department of Pathology, Wayne State University and Barbara Ann Karmanos Cancer Institute, Detroit, MI, USA
| | - Jing Li
- Metabolomics Core, Barbara Ann Karmanos Cancer Institute and Wayne State University, Detroit, MI, USA
| | - Jun Jiang
- Metabolomics Core, Barbara Ann Karmanos Cancer Institute and Wayne State University, Detroit, MI, USA
| | - Amy Tang
- Department of Public Health Services, Henry Ford Health System, Detroit, MI, USA
| | - Thomas Buekers
- Division of Gynecology Oncology, Department of Women's Health Services, Henry Ford Cancer Institute and Henry Ford Health System, Detroit, MI, USA; Department of Gynecology Oncology, Barbara Ann Karmanos Cancer Institute and Wayne State University, Detroit, MI, USA
| | - Robert Morris
- Department of Gynecology Oncology, Barbara Ann Karmanos Cancer Institute and Wayne State University, Detroit, MI, USA
| | - Adnan Munkarah
- Division of Gynecology Oncology, Department of Women's Health Services, Henry Ford Cancer Institute and Henry Ford Health System, Detroit, MI, USA
| | - Shailendra Giri
- Department of Neurology, Henry Ford Health System, Detroit, MI, USA
| | - Ramandeep Rattan
- Division of Gynecology Oncology, Department of Women's Health Services, Henry Ford Cancer Institute and Henry Ford Health System, Detroit, MI, USA; Department of Oncology, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
8
|
Kwon I, Song W, Jang Y, Choi MD, Vinci DM, Lee Y. Elevation of hepatic autophagy and antioxidative capacity by endurance exercise is associated with suppression of apoptosis in mice. Ann Hepatol 2021; 19:69-78. [PMID: 31611063 DOI: 10.1016/j.aohep.2019.08.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/16/2019] [Accepted: 08/18/2019] [Indexed: 02/04/2023]
Abstract
INTRODUCTION AND OBJECTIVES Endurance exercise (EXE) has emerged as a potent inducer of autophagy essential in maintaining cellular homeostasis in various tissues; however, the functional significance and molecular mechanisms of EXE-induced autophagy in the liver remain unclear. Thus, the aim of this study is to examine the signaling nexus of hepatic autophagy pathways occurring during acute EXE and a potential crosstalk between autophagy and apoptosis. MATERIALS AND METHODS C57BL/6 male mice were randomly assigned to sedentary control group (CON, n=9) and endurance exercise (EXE, n=9). Mice assigned to EXE were gradually acclimated to treadmill running and ran for 60min per day for five consecutive days. RESULTS Our data showed that EXE promoted hepatic autophagy via activation of canonical autophagy signaling pathways via mediating microtubule-associated protein B-light chain 3 II (LC3-II), autophagy protein 7 (ATG7), phosphorylated adenosine mono phosphate-activated protein kinase (p-AMPK), CATHEPSIN L, lysosome-associated membrane protein 2 (LAMP2), and a reduction in p62. Interestingly, this autophagy promotion concurred with enhanced anabolic activation via AKT-mammalian target of rapamycin (mTOR)-p70S6K signaling cascade and enhanced antioxidant capacity such as copper zinc superoxide dismutase (CuZnSOD), glutathione peroxidase (GPX), and peroxiredoxin 3 (PRX3), known to be as antagonists of autophagy. Moreover, exercise-induced autophagy was inversely related to apoptosis in the liver. CONCLUSIONS Our findings indicate that improved autophagy and antioxidant capacity, and potentiated anabolic signaling may be a potent non-pharmacological therapeutic strategy against diverse liver diseases.
Collapse
Affiliation(s)
- Insu Kwon
- Molecular and Cellular Exercise Physiology Laboratory, Department of Movement Sciences and Health, Usha Kundu, MD College of Health, University of West Florida, Pensacola, FLUSA
| | - Wankeun Song
- Molecular and Cellular Exercise Physiology Laboratory, Department of Movement Sciences and Health, Usha Kundu, MD College of Health, University of West Florida, Pensacola, FLUSA
| | - Yongchul Jang
- Molecular and Cellular Exercise Physiology Laboratory, Department of Movement Sciences and Health, Usha Kundu, MD College of Health, University of West Florida, Pensacola, FLUSA
| | - Myung D Choi
- Exercise Science, School of Health Sciences, Oakland University, Rochester, MIUSA
| | - Debra M Vinci
- Molecular and Cellular Exercise Physiology Laboratory, Department of Movement Sciences and Health, Usha Kundu, MD College of Health, University of West Florida, Pensacola, FLUSA
| | - Youngil Lee
- Molecular and Cellular Exercise Physiology Laboratory, Department of Movement Sciences and Health, Usha Kundu, MD College of Health, University of West Florida, Pensacola, FLUSA.
| |
Collapse
|
9
|
Benzarti M, Delbrouck C, Neises L, Kiweler N, Meiser J. Metabolic Potential of Cancer Cells in Context of the Metastatic Cascade. Cells 2020; 9:E2035. [PMID: 32899554 PMCID: PMC7563895 DOI: 10.3390/cells9092035] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/01/2020] [Accepted: 09/02/2020] [Indexed: 12/13/2022] Open
Abstract
The metastatic cascade is a highly plastic and dynamic process dominated by cellular heterogeneity and varying metabolic requirements. During this cascade, the three major metabolic pillars, namely biosynthesis, RedOx balance, and bioenergetics, have variable importance. Biosynthesis has superior significance during the proliferation-dominated steps of primary tumour growth and secondary macrometastasis formation and only minor relevance during the growth-independent processes of invasion and dissemination. Consequently, RedOx homeostasis and bioenergetics emerge as conceivable metabolic key determinants in cancer cells that disseminate from the primary tumour. Within this review, we summarise our current understanding on how cancer cells adjust their metabolism in the context of different microenvironments along the metastatic cascade. With the example of one-carbon metabolism, we establish a conceptual view on how the same metabolic pathway can be exploited in different ways depending on the current cellular needs during metastatic progression.
Collapse
Affiliation(s)
- Mohaned Benzarti
- Cancer Metabolism Group, Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg; (M.B.); (C.D.); (L.N.); (N.K.)
- Faculty of Science, Technology and Medicine, University of Luxembourg, 2 Avenue de l’Université, L-4365 Esch-sur-Alzette, Luxembourg
| | - Catherine Delbrouck
- Cancer Metabolism Group, Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg; (M.B.); (C.D.); (L.N.); (N.K.)
- Faculty of Science, Technology and Medicine, University of Luxembourg, 2 Avenue de l’Université, L-4365 Esch-sur-Alzette, Luxembourg
| | - Laura Neises
- Cancer Metabolism Group, Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg; (M.B.); (C.D.); (L.N.); (N.K.)
| | - Nicole Kiweler
- Cancer Metabolism Group, Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg; (M.B.); (C.D.); (L.N.); (N.K.)
| | - Johannes Meiser
- Cancer Metabolism Group, Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg; (M.B.); (C.D.); (L.N.); (N.K.)
| |
Collapse
|
10
|
Neitzel C, Demuth P, Wittmann S, Fahrer J. Targeting Altered Energy Metabolism in Colorectal Cancer: Oncogenic Reprogramming, the Central Role of the TCA Cycle and Therapeutic Opportunities. Cancers (Basel) 2020; 12:E1731. [PMID: 32610612 PMCID: PMC7408264 DOI: 10.3390/cancers12071731] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/24/2020] [Accepted: 06/25/2020] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC) is among the most frequent cancer entities worldwide. Multiple factors are causally associated with CRC development, such as genetic and epigenetic alterations, inflammatory bowel disease, lifestyle and dietary factors. During malignant transformation, the cellular energy metabolism is reprogrammed in order to promote cancer cell growth and proliferation. In this review, we first describe the main alterations of the energy metabolism found in CRC, revealing the critical impact of oncogenic signaling and driver mutations in key metabolic enzymes. Then, the central role of mitochondria and the tricarboxylic acid (TCA) cycle in this process is highlighted, also considering the metabolic crosstalk between tumor and stromal cells in the tumor microenvironment. The identified cancer-specific metabolic transformations provided new therapeutic targets for the development of small molecule inhibitors. Promising agents are in clinical trials and are directed against enzymes of the TCA cycle, including isocitrate dehydrogenase, pyruvate dehydrogenase kinase, pyruvate dehydrogenase complex (PDC) and α-ketoglutarate dehydrogenase (KGDH). Finally, we focus on the α-lipoic acid derivative CPI-613, an inhibitor of both PDC and KGDH, and delineate its anti-tumor effects for targeted therapy.
Collapse
Affiliation(s)
| | | | | | - Jörg Fahrer
- Division of Food Chemistry and Toxicology, Department of Chemistry, Technical University of Kaiserslautern, 67663 Kaiserslautern, Germany; (C.N.); (P.D.); (S.W.)
| |
Collapse
|
11
|
Li QJ, Wang ZG, Xie Y, Liu Q, Hu HL, Gao YX. Mechanistic evaluation of gastro-protective effects of KangFuXinYe on indomethacin-induced gastric damage in rats. Chin J Nat Med 2020; 18:47-56. [PMID: 31955823 DOI: 10.1016/s1875-5364(20)30004-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Indexed: 01/30/2023]
Abstract
KangFuXinYe (KFX), the ethanol extract of the dried whole body of Periplaneta americana, is a well-known important Chinese medicine preparation that has been used to treat digestive diseases such as gastric ulcers for many years in China. However, its therapeutic effect and mechanism are not yet well understood. Thus, the aim of this study was to investigate the gastro-protective effects of KangFuXinYe (KFX) in indomethacin-induced gastric damage. Rats were randomly divided into six groups as follows: control, treated with indomethacin (35 mg·kg-1), different dosages of KFX (2.57, 5.14 and 10.28 mL·kg-1, respectively) plus indomethacin, and sucralfate (1.71 mL·kg-1) plus indomethacin. After treatment, rat serum, stomach and gastric homogenates were collected for biochemical tests and examination of histopathology firstly. Rat serum was further used for metabolomics analysis to research possible mechanisms. Our results showed that KFX treatment alleviated indomethacin-induced histopathologic damage in rat gastric mucosa. Meanwhile, its treatment significantly increased cyclooxygenase-1 (COX-1), prostaglandin E2 (PGE2) and epidermal growth factor (EGF) levels in rat serum and gastric mucosa. Moreover, KFX decreased cyclooxygenase-2 (COX-2) and interleukin-6 (IL-6) levels. Nine metabolites were identified which intensities significantly changed in gastric damage rats, including 5-hydroxyindoleacetic acid, indoxylsulfuric acid, indolelactic acid, 4-hydroxyindole, pantothenic acid, isobutyryl carnitine, 3-methyl-2-oxovaleric acid, sphingosine 1-phosphate, and indometacin. These metabolic deviations came to closer to normal levels after KFX intervention. The results indicate that KFX (10.28 mL·kg-1) exerts protective effects on indomethacin-induced gastric damage by possible mechanisms of action (regulating tryptophan metabolism, protecting the mitochondria, and adjusting lipid metabolism, and reducing excessive indomethacin).
Collapse
Affiliation(s)
- Qi-Juan Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Zhan-Guo Wang
- Metabonomics Synergy Innovation Laboratory, School of Medicine and Nursing, Chengdu University, Chengdu 610106, China
| | - Yu Xie
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Qiao Liu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Hui-Ling Hu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Yong-Xiang Gao
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
12
|
Pohanka M. Antidotes Against Methanol Poisoning: A Review. Mini Rev Med Chem 2019; 19:1126-1133. [PMID: 30864518 DOI: 10.2174/1389557519666190312150407] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 02/20/2019] [Accepted: 03/06/2019] [Indexed: 12/12/2022]
Abstract
Methanol is the simplest alcohol. Compared to ethanol that is fully detoxified by metabolism. Methanol gets activated in toxic products by the enzymes, alcohol dehydrogenase and aldehyde dehydrogenase. Paradoxically, the same enzymes convert ethanol to harmless acetic acid. This review is focused on a discussion and overview of the literature devoted to methanol toxicology and antidotal therapy. Regarding the antidotal therapy, three main approaches are presented in the text: 1) ethanol as a competitive inhibitor in alcohol dehydrogenase; 2) use of drugs like fomepizole inhibiting alcohol dehydrogenase; 3) tetrahydrofolic acid and its analogues reacting with the formate as a final product of methanol metabolism. All the types of antidotal therapies are described and how they protect from toxic sequelae of methanol is explained.
Collapse
Affiliation(s)
- Miroslav Pohanka
- Faculty of Military Health Sciences, University of Defense, Trebesska 1575, Hradec Kralove CZ-50001, Czech Republic
| |
Collapse
|
13
|
Liu J, Lu W, Shi B, Klein S, Su X. Peroxisomal regulation of redox homeostasis and adipocyte metabolism. Redox Biol 2019; 24:101167. [PMID: 30921635 PMCID: PMC6434164 DOI: 10.1016/j.redox.2019.101167] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 03/01/2019] [Accepted: 03/10/2019] [Indexed: 12/26/2022] Open
Abstract
Peroxisomes are ubiquitous cellular organelles required for specific pathways of fatty acid oxidation and lipid synthesis, and until recently their functions in adipocytes have not been well appreciated. Importantly, peroxisomes host many oxygen-consumption reactions and play a major role in generation and detoxification of reactive oxygen species (ROS) and reactive nitrogen species (RNS), influencing whole cell redox status. Here, we review recent progress in peroxisomal functions in lipid metabolism as related to ROS/RNS metabolism and discuss the roles of peroxisomal redox homeostasis in adipogenesis and adipocyte metabolism. We provide a framework for understanding redox regulation of peroxisomal functions in adipocytes together with testable hypotheses for developing therapies for obesity and the related metabolic diseases.
Collapse
Affiliation(s)
- Jingjing Liu
- Department of Biochemistry and Molecular Biology, Soochow University College of Medicine, Suzhou, 215123, China
| | - Wen Lu
- Department of Biochemistry and Molecular Biology, Soochow University College of Medicine, Suzhou, 215123, China; Department of Endocrinology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Bimin Shi
- Department of Endocrinology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Samuel Klein
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Xiong Su
- Department of Biochemistry and Molecular Biology, Soochow University College of Medicine, Suzhou, 215123, China; Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
14
|
Chalker J, Gardiner D, Kuksal N, Mailloux RJ. Characterization of the impact of glutaredoxin-2 (GRX2) deficiency on superoxide/hydrogen peroxide release from cardiac and liver mitochondria. Redox Biol 2018; 15:216-227. [PMID: 29274570 PMCID: PMC5773472 DOI: 10.1016/j.redox.2017.12.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 12/07/2017] [Accepted: 12/13/2017] [Indexed: 01/30/2023] Open
Abstract
Mitochondria are critical sources of hydrogen peroxide (H2O2), an important secondary messenger in mammalian cells. Recent work has shown that O2•-/H2O2 emission from individual sites of production in mitochondria is regulated by protein S-glutathionylation. Here, we conducted the first examination of O2•-/H2O2 release rates from cardiac and liver mitochondria isolated from mice deficient for glutaredoxin-2 (GRX2), a matrix-associated thiol oxidoreductase that facilitates the S-glutathionylation and deglutathionylation of proteins. Liver mitochondria isolated from mice heterozygous (GRX2+/-) and homozygous (GRX2-/-) for glutaredoxin-2 displayed a significant decrease in O2•-/H2O2 release when oxidizing pyruvate or 2-oxoglutarate. The genetic deletion of the Grx2 gene was associated with increased protein expression of pyruvate dehydrogenase (PDH) but not 2-oxoglutarate dehydrogenase (OGDH). By contrast, O2•-/H2O2 production was augmented in cardiac mitochondria from GRX2+/- and GRX2-/- mice metabolizing pyruvate or 2-oxoglutarate which was associated with decreased PDH and OGDH protein levels. ROS production was augmented in liver and cardiac mitochondria metabolizing succinate. Inhibitor studies revealed that OGDH and Complex III served as high capacity ROS release sites in liver mitochondria. By contrast, Complex I and Complex III were found to be the chief O2•-/H2O2 emitters in cardiac mitochondria. These findings identify an essential role for GRX2 in regulating O2•-/H2O2 release from mitochondria in liver and cardiac tissue. Our results demonstrate that the GRX2-mediated regulation of O2•-/H2O2 release through the S-glutathionylation of mitochondrial proteins may play an integral role in controlling cellular ROS signaling.
Collapse
Affiliation(s)
- Julia Chalker
- Memorial University of Newfoundland, Department of Biochemistry, St. John's, Newfoundland, Canada
| | - Danielle Gardiner
- Memorial University of Newfoundland, Department of Biochemistry, St. John's, Newfoundland, Canada
| | - Nidhi Kuksal
- Memorial University of Newfoundland, Department of Biochemistry, St. John's, Newfoundland, Canada
| | - Ryan J Mailloux
- Memorial University of Newfoundland, Department of Biochemistry, St. John's, Newfoundland, Canada.
| |
Collapse
|
15
|
Mailloux RJ, Young A, O'Brien M, Gill RM. Simultaneous Measurement of Superoxide/Hydrogen Peroxide and NADH Production by Flavin-containing Mitochondrial Dehydrogenases. J Vis Exp 2018. [PMID: 29553554 DOI: 10.3791/56975] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
It has been reported that mitochondria can contain up to 12 enzymatic sources of reactive oxygen species (ROS). A majority of these sites include flavin-dependent respiratory complexes and dehydrogenases that produce a mixture of superoxide (O2●-) and hydrogen peroxide (H2O2). Accurate quantification of the ROS-producing potential of individual sites in isolated mitochondria can be challenging due to the presence of antioxidant defense systems and side reactions that also form O2●-/H2O2. Use of nonspecific inhibitors that can disrupt mitochondrial bioenergetics can also compromise measurements by altering ROS release from other sites of production. Here, we present an easy method for the simultaneous measurement of H2O2 release and nicotinamide adenine dinucleotide (NADH) production by purified flavin-linked dehydrogenases. For our purposes here, we have used purified pyruvate dehydrogenase complex (PDHC) and α-ketoglutarate dehydrogenase complex (KGDHC) of porcine heart origin as examples. This method allows for an accurate measure of native H2O2 release rates by individual sites of production by eliminating other potential sources of ROS and antioxidant systems. In addition, this method allows for a direct comparison of the relationship between H2O2 release and enzyme activity and the screening of the effectiveness and selectivity of inhibitors for ROS production. Overall, this approach can allow for the in-depth assessment of native rates of ROS release for individual enzymes prior to conducting more sophisticated experiments with isolated mitochondria or permeabilized muscle fiber.
Collapse
Affiliation(s)
- Ryan J Mailloux
- Department of Biochemistry, Memorial University of Newfoundland;
| | - Adrian Young
- Department of Biochemistry, Memorial University of Newfoundland
| | - Marisa O'Brien
- Department of Biochemistry, Memorial University of Newfoundland
| | | |
Collapse
|