1
|
Kollmar J, Xu J, Gonzalves D, Baur JA, Li LZ, Tchou J, Xu HN. Differential Mitochondrial Redox Responses to the Inhibition of NAD + Salvage Pathway of Triple Negative Breast Cancer Cells. Cancers (Basel) 2024; 17:7. [PMID: 39796638 PMCID: PMC11718843 DOI: 10.3390/cancers17010007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 12/15/2024] [Accepted: 12/18/2024] [Indexed: 01/13/2025] Open
Abstract
Background/Objectives: Cancer cells rely on metabolic reprogramming that is supported by altered mitochondrial redox status and an increased demand for NAD+. Over expression of Nampt, the rate-limiting enzyme of the NAD+ biosynthesis salvage pathway, is common in breast cancer cells, and more so in triple negative breast cancer (TNBC) cells. Targeting the salvage pathway has been pursued for cancer therapy. However, TNBC cells have heterogeneous responses to Nampt inhibition, which contributes to the diverse outcomes. There is a lack of imaging biomarkers to differentiate among TNBC cells under metabolic stress and identify which are responsive. We aimed to characterize and differentiate among a panel of TNBC cell lines under NAD-deficient stress and identify which subtypes are more dependent on the NAD salvage pathway. Methods: Optical redox imaging (ORI), a label-free live cell imaging microscopy technique was utilized to acquire intrinsic fluorescence intensities of NADH and FAD-containing flavoproteins (Fp) thus the mitochondrial redox ratio Fp/(NADH + Fp) in a panel of TNBC cell lines. Various fluorescence probes were then added to the cultures to image the mitochondrial ROS, mitochondrial membrane potential, mitochondrial mass, and cell number. Results: Various TNBC subtypes are sensitive to Nampt inhibition in a dose- and time-dependent manner, they have differential mitochondrial redox responses; furthermore, the mitochondrial redox indices linearly correlated with mitochondrial ROS induced by various doses of a Nampt inhibitor. Moreover, the changes in the redox indices correlated with growth inhibition. Additionally, the redox state was found fully recovered after removing the Nampt inhibitor. Conclusions: This study supports the utility of ORI in rapid metabolic phenotyping of TNBC cells under NAD-deficient stress to identify responsive cells and biomarkers of treatment response, facilitating combination therapy strategies.
Collapse
Affiliation(s)
- Jack Kollmar
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.K.); (D.G.); (J.T.)
| | - Junmei Xu
- Britton Chance Laboratory of Redox Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.X.); (L.Z.L.)
| | - Diego Gonzalves
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.K.); (D.G.); (J.T.)
| | - Joseph A. Baur
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Lin Z. Li
- Britton Chance Laboratory of Redox Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.X.); (L.Z.L.)
| | - Julia Tchou
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.K.); (D.G.); (J.T.)
| | - He N. Xu
- Britton Chance Laboratory of Redox Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.X.); (L.Z.L.)
| |
Collapse
|
2
|
Modica-Napolitano JS, Murray M, Thibault J, Haley-Read JP, Nixdorf L, Shanahan B, Iacovella N, Reyes C. The In Vitro Cytotoxic Effect of Elesclomol on Breast Adenocarcinoma Cells Is Enhanced by Concurrent Treatment with Glycolytic Inhibitors. Cancers (Basel) 2024; 16:4054. [PMID: 39682240 DOI: 10.3390/cancers16234054] [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: 08/29/2024] [Revised: 11/18/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Background/Objectives: Glycolysis and mitochondrial oxidative phosphorylation are the two major metabolic pathways for cellular ATP production. The metabolic plasticity displayed by cancer cells allows them to effectively shift between each of these pathways as a means of adapting to various growth conditions, thus ensuring their survival, proliferation and disease progression. Metabolic plasticity also provides cancer cells with the ability to circumvent many traditional monotherapies aimed at only one or the other of the major ATP-producing pathways. The purpose of this study was to determine the effectiveness of a dual treatment strategy aimed simultaneously at both pathways of ATP production in human breast cancer cells. It was hypothesized that concurrent exposure of these cells to the mitochondria-targeting chemotherapeutic agent, elesclomol, in combination with either of two glycolytic inhibitors, 2-deoxy-D-glucose or 3-bromopyruvate, would yield greater in vitro anticancer effects than those observed for any of the compounds used as a single agent. Methods: Cytotoxicity and clonogenic assays were employed to assess the survival and proliferation of MCF7 and MDA-MB-231 human breast adenocarcinoma cells exposed to the compounds alone and in combination. Results: The data obtained show that the cancer-cell-killing and antiproliferative effects of the dual treatment were significantly enhanced compared to those observed for any of the compounds alone. Conclusions: The results of this study are important in that they suggest the possibility of a novel and effective chemotherapeutic strategy for breast cancer cell killing.
Collapse
Affiliation(s)
| | - Morgan Murray
- Department of Biology, Merrimack College, North Andover, MA 01845, USA
| | - Jacob Thibault
- Department of Biology, Merrimack College, North Andover, MA 01845, USA
| | | | - Lauren Nixdorf
- Department of Biology, Merrimack College, North Andover, MA 01845, USA
| | - Bridget Shanahan
- Department of Biology, Merrimack College, North Andover, MA 01845, USA
| | | | - Carlos Reyes
- Department of Biology, Merrimack College, North Andover, MA 01845, USA
| |
Collapse
|
3
|
Vozgirdaite D, Hervé-Aubert K, Uzbekov R, Chourpa I, Allard-Vannier E. Design, optimization, characterization, and in vitro evaluation of metformin-loaded liposomes for triple negative breast cancer treatment. J Liposome Res 2024; 34:547-561. [PMID: 38459750 DOI: 10.1080/08982104.2024.2321528] [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: 10/06/2023] [Revised: 01/22/2024] [Accepted: 02/16/2024] [Indexed: 03/10/2024]
Abstract
Recently, metformin (Met) has shown to have antineoplastic properties in cancer treatment by improving hypoxic tumor conditions, and causing reduction in the synthesis of biomolecules, which are vital for cancer growth. However, as an orally administered drug, Met has low bioavailability and rapid renal clearance. Thus, the goal of this study was to vectorize Met inside liposomes in the context of triple negative breast cancer (TNBC), which currently lacks treatment options when compared to other types of breast cancer. Vectorization of Met inside liposomes was done using Bangham method by implementing double design of experiment methodology to increase Met drug loading (minimum-run resolution V characterization design and Box-Behnken design), as it is generally extremely low for hydrophilic molecules. Optimization of Met-loaded liposome synthesis was successfully achieved with drug loading of 190 mg/g (19% w/w). The optimal Met-liposomes were 170 nm in diameter with low PdI (< 0.1) and negative surface charge (-20 mV), exhibiting sustained Met release at pH 7.4. The liposomal Met delivery system was stable over several months, and successfully reduced TNBC cell proliferation due to the encapsulated drug. This study is one the first reports addressing liposome formulation through thin-film hydration using two design of experiment methods aiming to increase drug loading of Met.
Collapse
Affiliation(s)
- Daiva Vozgirdaite
- UPR 4301 CBM, CNRS, NMNS department, University of Tours, Tours, France
| | | | - Rustem Uzbekov
- Laboratoire Biologie Cellulaire et Microscopie Electronique, Faculty of Medicine, University of Tours, Tours, France
- Faculty of Bioengineering and Bioinformatics, Moscow State University, Moscow, Russia
| | - Igor Chourpa
- UPR 4301 CBM, CNRS, NMNS department, University of Tours, Tours, France
| | | |
Collapse
|
4
|
Rossi T, Iorio E, Chirico M, Pisanu ME, Amodio N, Cantafio MEG, Perrotta I, Colciaghi F, Fiorillo M, Gianferrari A, Puccio N, Neri A, Ciarrocchi A, Pistoni M. BET inhibitors (BETi) influence oxidative phosphorylation metabolism by affecting mitochondrial dynamics leading to alterations in apoptotic pathways in triple-negative breast cancer (TNBC) cells. Cell Prolif 2024; 57:e13730. [PMID: 39223828 PMCID: PMC11628750 DOI: 10.1111/cpr.13730] [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] [Received: 01/25/2024] [Revised: 07/04/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024] Open
Abstract
Repressing BET proteins' function using bromodomain inhibitors (BETi) has been shown to elicit antitumor effects by regulating the transcription of genes downstream of BRD4. We previously showed that BETi promoted cell death of triple-negative breast cancer (TNBC) cells. Here, we proved that BETi induce altered mitochondrial dynamics fitness in TNBC cells falling in cell death. We demonstrated that BETi treatment downregulated the expression of BCL-2, and proteins involved in mitochondrial fission and increased fused mitochondria. Impaired mitochondrial fission affected oxidative phosphorylation (OXPHOS) inducing the expression of OXPHOS-related genes, SDHa and ATP5a, and increased cell death. Consistently, the amount of mitochondrial DNA and mitochondrial membrane potential (∆Ψm) increased in BETi-treated cells compared to control cells. Lastly, BETi in combination with Metformin reduced cell growth. Our results indicate that mitochondrial dynamics and OXPHOS metabolism support breast cancer proliferation and represent novel BETi downstream targets in TNBC cells.
Collapse
Affiliation(s)
- Teresa Rossi
- Laboratory of Translational ResearchAUSL‐IRCCS di Reggio EmiliaReggio EmilaItaly
| | - Egidio Iorio
- High Resolution NMR UnitCore Facilities, Istituto Superiore di SanitàRomeItaly
| | - Mattea Chirico
- High Resolution NMR UnitCore Facilities, Istituto Superiore di SanitàRomeItaly
| | - Maria Elena Pisanu
- High Resolution NMR UnitCore Facilities, Istituto Superiore di SanitàRomeItaly
| | - Nicola Amodio
- Department of Experimental and Clinical MedicineUniversity Magna Graecia of CatanzaroCatanzaroItaly
| | | | - Ida Perrotta
- Department of Biology, Ecology and Earth SciencesCentre for Microscopy and Microanalysis (CM2), University of CalabriaCosenzaItaly
| | | | - Marco Fiorillo
- Department of Pharmacy, Health and Nutritional SciencesUniversity of CalabriaRendeItaly
| | - Alessia Gianferrari
- Laboratory of Translational ResearchAUSL‐IRCCS di Reggio EmiliaReggio EmilaItaly
| | - Noemi Puccio
- Laboratory of Translational ResearchAUSL‐IRCCS di Reggio EmiliaReggio EmilaItaly
| | - Antonino Neri
- Scientific DirectorateAUSL‐IRCCS di Reggio EmiliaReggio EmilaItaly
| | - Alessia Ciarrocchi
- Laboratory of Translational ResearchAUSL‐IRCCS di Reggio EmiliaReggio EmilaItaly
| | - Mariaelena Pistoni
- Laboratory of Translational ResearchAUSL‐IRCCS di Reggio EmiliaReggio EmilaItaly
| |
Collapse
|
5
|
Gong S, Huang R, Wang M, Lian F, Wang Q, Liao Z, Fan C. Comprehensive analysis of the metabolomics and transcriptomics uncovers the dysregulated network and potential biomarkers of Triple Negative Breast Cancer. J Transl Med 2024; 22:1016. [PMID: 39529035 PMCID: PMC11552364 DOI: 10.1186/s12967-024-05843-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is known for its aggressive nature, lack of effective diagnostic tools and treatments, and generally poor prognosis. The objective of this study was to investigate metabolic changes in TNBC using metabolomics approaches and explore the underlying mechanisms through integrated analysis with transcriptomics. In this study, serum untargeted metabolic profiles were first examined between 18 TNBC patients and 21 healthy control (HC) subjects using liquid chromatography-mass spectrometry (LC-MS), identifying a total of 22 significantly differential metabolites (DMs). Subsequently, receiver operating characteristic analysis revealed that 7-methylguanine could serve as a potential biomarker for TNBC in both the discovery and validation sets. Additionally, transcriptomic datasets were retrieved from the GEO database to identify differentially expressed genes (DEGs) between TNBC and normal tissues. An integrative analysis of the DMs and DEGs was conducted, uncovering potential molecular mechanisms underlying TNBC. Notably, three pathways-tyrosine metabolism, phenylalanine metabolism, and glycolysis/gluconeogenesis-were enriched, providing insight into the energy metabolism disorders in TNBC. Within these pathways, two DMs (4-hydroxyphenylacetaldehyde and oxaloacetic acid) and six DEGs (MAOA, ADH1B, ADH1C, AOC3, TAT, and PCK1) were identified as key components. In summary, this study highlights metabolic biomarkers that could potentially be used for the diagnosis and screening of TNBC. The comprehensive analysis of metabolomics and transcriptomics data offers a validated and in-depth understanding of TNBC metabolism.
Collapse
Affiliation(s)
- Sisi Gong
- The Clinical Laboratory Center, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, P.R. China
| | - Rongfu Huang
- The Clinical Laboratory Center, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, P.R. China
| | - Meie Wang
- The Clinical Laboratory Center, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, P.R. China
| | - Fen Lian
- The Clinical Laboratory Center, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, P.R. China
| | - Qingshui Wang
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, P.R. China.
| | - Zhijun Liao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, P.R. China.
| | - Chunmei Fan
- The Clinical Laboratory Center, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, P.R. China.
| |
Collapse
|
6
|
Orobator ON, Mertens RT, Obisesan OA, Awuah SG. Energy and endoplasmic reticulum stress induction by gold(III) dithiocarbamate and 2-deoxyglucose synergistically trigger cell death in breast cancer. J Biol Chem 2024:107949. [PMID: 39481597 DOI: 10.1016/j.jbc.2024.107949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/01/2024] [Accepted: 10/08/2024] [Indexed: 11/02/2024] Open
Abstract
The elusiveness of triple-negative breast cancer from targeted therapy has redirected focus towards exploiting the metabolic shortcomings of these highly metastatic subtypes of breast cancer. Cueing from the metabolic heterogeneity of TNBC and the exposition of the dual dependence of some TNBCs on OXPHOS and glycolysis for ATP, we herein report the efficacy of cotreatment of TNBCs with an OXPHOS inhibitor, 2a and 2DG, a potent glycolysis inhibitor. 2a-2DG cotreatment inhibited TNBC cell proliferation with IC50 of ∼5 to 36 times lower than that of 2a alone and over 5000 times lower than IC50 of 2DG alone. 2a-2DG cotreatment suppressed mitochondrial ATP production and significantly induced AMPK activation. Mechanistic studies revealed the distinct yet synergistic contributions of 2a and 2DG to the antiproliferative effect of the cotreatment. While 2a induced apoptotic cell death, 2DG sensitized TNBCs to the antiproliferative effects of 2a via endoplasmic reticulum stress induction. Strikingly, the combination of 2a-2DG ablated SUM159 tumors in an orthotopic xenograft mouse model. This study highlights the synergistic effect of a gold-based complex with 2DG and the potential benefit of multi-metabolic pathways targeting as an effective therapeutic strategy against TNBCs.
Collapse
Affiliation(s)
- Owamagbe N Orobator
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - R Tyler Mertens
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Oluwatosin A Obisesan
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Samuel G Awuah
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States; Center for Pharmaceutical Research and Innovation, Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536, USA; Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40536, USA; Center for Bioelectronics and Nanomedicine, University of Kentucky, Lexington KY, 40506, USA.
| |
Collapse
|
7
|
Khan A, Rehman AU, Siddiqui S, Khan J, Massey S, Singh P, Saluja D, Husain SA, Iqbal MA. Withaferin A decreases glycolytic reprogramming in breast cancer. Sci Rep 2024; 14:23147. [PMID: 39366987 PMCID: PMC11452501 DOI: 10.1038/s41598-024-72221-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 09/04/2024] [Indexed: 10/06/2024] Open
Abstract
Reprogrammed glucose metabolism is considered as the hallmark of cancer with therapeutic implications. Phytocompounds have potential to inhibit cancer metabolism. Here, we tested the ability of Withaferin A (WA), a withanolide derived from Withania somnifera, in modulating cancer metabolism. The assessed effect of WA on aerobic glycolysis in breast cancer cell lines showed that WA decreases the glucose uptake, lactate production and ATP generation by inhibiting the expression of key glycolytic enzymes i.e., GLUT1, HK2 and PKM2. We also identified that WA induced inhibition of cancer glycolysis by targeting c-myc as validated by silencing experiments followed by metabolic readouts. Decreased glycolysis resulted in reduced cell viability, biomass and colony forming ability of breast cancer cells. To further validate our in vitro findings in breast cancer patients, we analyzed 90 metabolic pathways in ~ 2000 breast tumors and observed that glycolysis is the most deregulated pathway in breast tumors. Deregulated glycolysis also predicted poor prognosis in breast cancer patients. In addition, patient data showed correlation between c-myc expression and glycolytic deregulation in breast cancer. Taken together, our results highlight the role of WA in inhibiting breast cancer metabolism via c-myc/glycolysis axis.
Collapse
Affiliation(s)
- Asifa Khan
- Department of Biosciences, Jamia Millia Islamia (A Central University), New Delhi, 110025, India
| | - Asad Ur Rehman
- Medical Biotechnology Laboratory, Dr B R Ambedkar Center for Biomedical Research, University of Delhi, New Delhi, India
| | - Shumaila Siddiqui
- Division of Cancer Biology, CSIR-Central Drug Research Institute, Lucknow, UP, India
| | - Jiyauddin Khan
- Medical Biotechnology Laboratory, Dr B R Ambedkar Center for Biomedical Research, University of Delhi, New Delhi, India
| | - Sheersh Massey
- Department of Biosciences, Jamia Millia Islamia (A Central University), New Delhi, 110025, India
| | - Prithvi Singh
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia (A Central University), New Delhi, 110025, India
| | - Daman Saluja
- Medical Biotechnology Laboratory, Dr B R Ambedkar Center for Biomedical Research, University of Delhi, New Delhi, India
| | - Syed Akhtar Husain
- Department of Biosciences, Jamia Millia Islamia (A Central University), New Delhi, 110025, India.
| | - Mohammad Askandar Iqbal
- Department of Biotechnology, Jamia Millia Islamia (A Central University), New Delhi, 110025, India.
- Thumbay Research Institute for Precision Medicine (TRIPM), College of Medicine, Gulf Medical University, Ajman, United Arab Emirates.
| |
Collapse
|
8
|
Carlosama C, Arévalo C, Jimenez MC, Lasso P, Urueña C, Fiorentino S, Barreto A. Triple negative breast cancer migration is modified by mitochondrial metabolism alteration induced by natural extracts of C. spinosa and P. alliacea. Sci Rep 2024; 14:20253. [PMID: 39215068 PMCID: PMC11364553 DOI: 10.1038/s41598-024-70550-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024] Open
Abstract
Tumor metabolism is a crucial aspect of cancer development, and mitochondria plays a significant role in the aggressiveness and metastasis of tumors. As a result, mitochondria have become a promising therapeutic target in cancer treatment, leading to the development of compounds known as mitocans. In our group, we have consolidated the search of anticancer therapies based on natural products derived from plants, obtaining extracts such as P2Et from Caesalpinia spinosa and Anamu-SC from Petiveria alliacea, which have been shown to have antitumor activities in different cancer models. These extracts, due to their complex molecular composition, can interfere with multiple functions during tumor progression. To better understand how these natural products operate (P2Et and Anamu-SC), we constructed a model using 4T1 murine breast cancer cells with reduced expression of genes associated with glycolysis (Hexokinase-2) and mitochondrial function (Cqbp). The results indicate that the cells were more sensitive to the Anamu-SC extract, showing significant decreases in glucose consumption, ATP production, and oxygen consumption rate. Additionally, we observed changes in mitochondrial function, which reduced the cells' ability to migrate, particularly when C1qbp was silenced. This triple-negative breast cancer model allows us to identify potential natural products that can modulate tumor cell metabolism.
Collapse
Affiliation(s)
- Carolina Carlosama
- Grupo de Inmunobiología y Biología Celular, Unidad de Investigación en Ciencias Biomédicas, Facultad de Ciencias, Pontificia Universidad Javeriana, Carrera 7a. No. 43-82, Ed. 50, Lab. 101, 110211, Bogotá, Colombia
| | - Cindy Arévalo
- Grupo de Inmunobiología y Biología Celular, Unidad de Investigación en Ciencias Biomédicas, Facultad de Ciencias, Pontificia Universidad Javeriana, Carrera 7a. No. 43-82, Ed. 50, Lab. 101, 110211, Bogotá, Colombia
| | - María Camila Jimenez
- Grupo de Inmunobiología y Biología Celular, Unidad de Investigación en Ciencias Biomédicas, Facultad de Ciencias, Pontificia Universidad Javeriana, Carrera 7a. No. 43-82, Ed. 50, Lab. 101, 110211, Bogotá, Colombia
| | - Paola Lasso
- Grupo de Inmunobiología y Biología Celular, Unidad de Investigación en Ciencias Biomédicas, Facultad de Ciencias, Pontificia Universidad Javeriana, Carrera 7a. No. 43-82, Ed. 50, Lab. 101, 110211, Bogotá, Colombia
| | - Claudia Urueña
- Grupo de Inmunobiología y Biología Celular, Unidad de Investigación en Ciencias Biomédicas, Facultad de Ciencias, Pontificia Universidad Javeriana, Carrera 7a. No. 43-82, Ed. 50, Lab. 101, 110211, Bogotá, Colombia
| | - Susana Fiorentino
- Grupo de Inmunobiología y Biología Celular, Unidad de Investigación en Ciencias Biomédicas, Facultad de Ciencias, Pontificia Universidad Javeriana, Carrera 7a. No. 43-82, Ed. 50, Lab. 101, 110211, Bogotá, Colombia
| | - Alfonso Barreto
- Grupo de Inmunobiología y Biología Celular, Unidad de Investigación en Ciencias Biomédicas, Facultad de Ciencias, Pontificia Universidad Javeriana, Carrera 7a. No. 43-82, Ed. 50, Lab. 101, 110211, Bogotá, Colombia.
| |
Collapse
|
9
|
Ahn J, Lee JW, Nam SM, Kim DK, Cho SK, Choi HK. Integrative multi-omics analysis reveals ortho-topolin riboside exhibits anticancer activity by regulating metabolic pathways in radio-resistant triple negative breast cancer cells. Chem Biol Interact 2024; 398:111089. [PMID: 38823535 DOI: 10.1016/j.cbi.2024.111089] [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: 12/19/2023] [Revised: 04/07/2024] [Accepted: 05/29/2024] [Indexed: 06/03/2024]
Abstract
Radio-resistant triple negative breast cancer (TNBC) is resistant to conventional drugs and radiation therapy. ortho-topolin riboside (oTR) has been evaluated for its anticancer activity in several types of cancer cells. However, its anti-proliferative activity in radio-resistant TNBC cells has not yet been reported. Therefore, we investigated the anti-proliferative activity of oTR in radio-resistant TNBC cells, and performed metabolome, lipidome, transcriptome, and proteome profiling to reveal the mechanisms of the anticancer activity of oTR. oTR showed cytotoxicity against radio-resistant TNBC cells with an inhibitory concentration (IC50) value of 7.78 μM. Significantly decreased (p value < 0.05) basal and compensatory glycolysis were observed in the oTR-treated group than untreated group. Mitochondrial spare respiratory capacity, which is relevant to cell fitness and flexibility, was significantly decreased (p value < 0.05) in the oTR-treated group. The major metabolic pathways significantly altered by oTR according to metabolome, transcriptome, and proteome profiles were the glycerolipid/glycerophospholipid pathway (log2(FC) of MGLL = -0.13, log2(FC) of acylglycerol lipase = -1.35, log2(FC) of glycerol = -0.81), glycolysis (log2(FC) of EGLN1 = 0.16, log2(FC) of EGLN1 = 0.62, log2(FC) of glucose = -0.76, log2(FC) of lactate = -0.81), and kynurenine pathway (log2(FC) of KYNU = 0.29, log2(FC) of kynureninase = 0.55, log2(FC) of alanine = 0.72). Additionally, proline metabolism (log2(FC) of PYCR1 = -0.17, log2(FC) of proline = -0.73) was significantly altered in the metabolomic and transcriptomic profiles. The MAPK signaling pathway (log2(FC) of CCN1 = -0.15, log2(FC) of CCN family member 1 = -1.02) and Rap 1 signaling pathway (log2(FC) of PARD6B = -0.28, log2(FC) of PAR6B = -3.13) were also significantly altered in transcriptomic and proteomic profiles. The findings of this study revealed that oTR has anticancer activity in radio-resistant TNBC cells by affecting various metabolic pathways, suggesting the potential of oTR as a novel anticancer agent for radio-resistant TNBC patients.
Collapse
Affiliation(s)
- Junyoung Ahn
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Ji Won Lee
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Seung Min Nam
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Dae Kyeong Kim
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju, 63243, Republic of Korea
| | - Somi Kim Cho
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju, 63243, Republic of Korea; Subtropical/Tropical Organism Gene Bank, Jeju National University, Jeju, 63243, Republic of Korea.
| | - Hyung-Kyoon Choi
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea.
| |
Collapse
|
10
|
Pires ICB, Shuchi SI, Tostes BDVA, Santos DKDDN, Burnett WL, Leonce BC, Harvey OR, Coffer JL, de Sousa Filho IA, de Athayde-Filho PF, Junior SA, Mathis JM. Theranostics Using MCM-41-Based Mesoporous Silica Nanoparticles: Integrating Magnetic Resonance Imaging and Novel Chemotherapy for Breast Cancer Treatment. Int J Mol Sci 2024; 25:8097. [PMID: 39125669 PMCID: PMC11311303 DOI: 10.3390/ijms25158097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/11/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Advanced breast cancer remains a significant oncological challenge, requiring new approaches to improve clinical outcomes. This study investigated an innovative theranostic agent using the MCM-41-NH2-DTPA-Gd3⁺-MIH nanomaterial, which combined MRI imaging for detection and a novel chemotherapy agent (MIH 2.4Bl) for treatment. The nanomaterial was based on the mesoporous silica type, MCM-41, and was optimized for drug delivery via functionalization with amine groups and conjugation with DTPA and complexation with Gd3+. MRI sensitivity was enhanced by using gadolinium-based contrast agents, which are crucial in identifying early neoplastic lesions. MIH 2.4Bl, with its unique mesoionic structure, allows effective interactions with biomolecules that facilitate its intracellular antitumoral activity. Physicochemical characterization confirmed the nanomaterial synthesis and effective drug incorporation, with 15% of MIH 2.4Bl being adsorbed. Drug release assays indicated that approximately 50% was released within 8 h. MRI phantom studies demonstrated the superior imaging capability of the nanomaterial, with a relaxivity significantly higher than that of the commercial agent Magnevist. In vitro cellular cytotoxicity assays, the effectiveness of the nanomaterial in killing MDA-MB-231 breast cancer cells was demonstrated at an EC50 concentration of 12.6 mg/mL compared to an EC50 concentration of 68.9 mg/mL in normal human mammary epithelial cells (HMECs). In vivo, MRI evaluation in a 4T1 syngeneic mouse model confirmed its efficacy as a contrast agent. This study highlighted the theranostic capabilities of MCM-41-NH2-DTPA-Gd3⁺-MIH and its potential to enhance breast cancer management.
Collapse
Affiliation(s)
- Indira C. B. Pires
- Department of Chemistry, Federal University of Pernambuco, Recife 50670-901, PE, Brazil; (I.C.B.P.); (B.d.V.A.T.); (D.K.D.d.N.S.)
| | - Samia I. Shuchi
- School of Biomedical Sciences, Departments of Microbiology, Immunology, and Genetics and Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA;
| | - Braulio de V. A. Tostes
- Department of Chemistry, Federal University of Pernambuco, Recife 50670-901, PE, Brazil; (I.C.B.P.); (B.d.V.A.T.); (D.K.D.d.N.S.)
| | - Dayane K. D. do N. Santos
- Department of Chemistry, Federal University of Pernambuco, Recife 50670-901, PE, Brazil; (I.C.B.P.); (B.d.V.A.T.); (D.K.D.d.N.S.)
| | - William L. Burnett
- Department of Chemistry and Biochemistry, Texas Christian University, Fort Worth, TX 76109, USA (B.C.L.); (O.R.H.); (J.L.C.)
| | - Burke C. Leonce
- Department of Chemistry and Biochemistry, Texas Christian University, Fort Worth, TX 76109, USA (B.C.L.); (O.R.H.); (J.L.C.)
| | - Omar R. Harvey
- Department of Chemistry and Biochemistry, Texas Christian University, Fort Worth, TX 76109, USA (B.C.L.); (O.R.H.); (J.L.C.)
| | - Jeffery L. Coffer
- Department of Chemistry and Biochemistry, Texas Christian University, Fort Worth, TX 76109, USA (B.C.L.); (O.R.H.); (J.L.C.)
| | - Idio Alves de Sousa Filho
- Institute of Chemistry, Federal Rural University of Rio de Janeiro, Rio de Janeiro 23890-000, RJ, Brazil;
| | | | - Severino A. Junior
- Department of Chemistry, Federal University of Pernambuco, Recife 50670-901, PE, Brazil; (I.C.B.P.); (B.d.V.A.T.); (D.K.D.d.N.S.)
| | - J. Michael Mathis
- School of Biomedical Sciences, Departments of Microbiology, Immunology, and Genetics and Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA;
| |
Collapse
|
11
|
Manjunath M, Ravindran F, Sharma S, Siddiqua H, Raghavan SC, Choudhary B. Disarib, a Specific BCL2 Inhibitor, Induces Apoptosis in Triple-Negative Breast Cancer Cells and Impedes Tumour Progression in Xenografts by Altering Mitochondria-Associated Processes. Int J Mol Sci 2024; 25:6485. [PMID: 38928195 PMCID: PMC11203414 DOI: 10.3390/ijms25126485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
Targeted cancer therapy aims to disrupt the functions of proteins that regulate cancer progression, mainly by using small molecule inhibitors (SMIs). SMIs exert their effect by modulating signalling pathways, organelle integrity, chromatin components, and several biosynthetic processes essential for cell division and survival. Antiapoptotic protein BCL2 is highly upregulated in many cancers compared with normal cells, making it an ideal target for cancer therapy. Around 75% of primary breast cancers overexpress BCL2, providing an opportunity to explore BCL2 inhibitors as a therapeutic option. Disarib is an SMI that has been developed as a selective BCL2 inhibitor. Disarib works by disrupting BCL2-BAK interaction and activating intrinsic apoptotic pathways in leukemic cells while sparing normal cells. We investigated the effects of Disarib, a BCL2 specific inhibitor, on breast cancer cells and xenografts. Cytotoxicity and fluorometric assays revealed that Disarib induced cell death by increasing reactive oxygen species and activating intrinsic apoptotic pathways in Triple-Negative Breast Cancer cells (MDA-MB-231 and MDA-MB-468). Disarib also affected the colony-forming properties of these cells. MDA-MB-231- and MDA-MB-468-derived xenografts showed a significant reduction in tumours upon Disarib treatment. Through the transcriptomics approach, we also explored the influence of BCL2 inhibitors on energy metabolism, mitochondrial dynamics, and epithelial-to-mesenchymal transition (EMT). Mitochondrial dynamics and glucose metabolism mainly regulate energy metabolism. The change in energetics regulates tumour growth through epithelial-mesenchymal transition, and angiogenesis. RNA sequencing (RNAseq) analysis revealed that BCL2 inhibitors ABT-199 and Disarib maintain Oxphos levels in MDA-MB-231. However, key glycolytic genes were significantly downregulated. Mitochondrial fission genes were seen to be downregulated both in RNAseq data and semi quantitative real time polymerase chain reaction (qRTPCR) in Disarib-treated TNBC cells and xenografts. Lastly, Disarib inhibited wound healing and epithelial-to-mesenchymal transition. This study showed that Disarib disrupts mitochondrial function, activates the intrinsic apoptotic pathway in breast cancer, and inhibits epithelial-to-mesenchymal transition both in vitro and in vivo. These findings highlight Disarib's potential as a multifaceted therapeutic strategy for patients with Triple-Negative Breast Cancer.
Collapse
Affiliation(s)
- Meghana Manjunath
- Department of Biotechnology and Applied Bioinformatics, Institute of Bioinformatics and Applied Biotechnology, Electronic City Phase 1, Bengaluru 560100, India
| | - Febina Ravindran
- Department of Biotechnology and Applied Bioinformatics, Institute of Bioinformatics and Applied Biotechnology, Electronic City Phase 1, Bengaluru 560100, India
| | - Shivangi Sharma
- Department of Biotechnology and Applied Bioinformatics, Institute of Bioinformatics and Applied Biotechnology, Electronic City Phase 1, Bengaluru 560100, India
- Indian Institute of Science, Bengaluru 560012, India; (H.S.); (S.C.R.)
| | - Humaira Siddiqua
- Indian Institute of Science, Bengaluru 560012, India; (H.S.); (S.C.R.)
| | | | - Bibha Choudhary
- Department of Biotechnology and Applied Bioinformatics, Institute of Bioinformatics and Applied Biotechnology, Electronic City Phase 1, Bengaluru 560100, India
| |
Collapse
|
12
|
Das C, Bhattacharya A, Adhikari S, Mondal A, Mondal P, Adhikary S, Roy S, Ramos K, Yadav KK, Tainer JA, Pandita TK. A prismatic view of the epigenetic-metabolic regulatory axis in breast cancer therapy resistance. Oncogene 2024; 43:1727-1741. [PMID: 38719949 PMCID: PMC11161412 DOI: 10.1038/s41388-024-03054-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 06/09/2024]
Abstract
Epigenetic regulation established during development to maintain patterns of transcriptional expression and silencing for metabolism and other fundamental cell processes can be reprogrammed in cancer, providing a molecular mechanism for persistent alterations in phenotype. Metabolic deregulation and reprogramming are thus an emerging hallmark of cancer with opportunities for molecular classification as a critical preliminary step for precision therapeutic intervention. Yet, acquisition of therapy resistance against most conventional treatment regimens coupled with tumor relapse, continue to pose unsolved problems for precision healthcare, as exemplified in breast cancer where existing data informs both cancer genotype and phenotype. Furthermore, epigenetic reprograming of the metabolic milieu of cancer cells is among the most crucial determinants of therapeutic resistance and cancer relapse. Importantly, subtype-specific epigenetic-metabolic interplay profoundly affects malignant transformation, resistance to chemotherapy, and response to targeted therapies. In this review, we therefore prismatically dissect interconnected epigenetic and metabolic regulatory pathways and then integrate them into an observable cancer metabolism-therapy-resistance axis that may inform clinical intervention. Optimally coupling genome-wide analysis with an understanding of metabolic elements, epigenetic reprogramming, and their integration by metabolic profiling may decode missing molecular mechanisms at the level of individual tumors. The proposed approach of linking metabolic biochemistry back to genotype, epigenetics, and phenotype for specific tumors and their microenvironment may thus enable successful mechanistic targeting of epigenetic modifiers and oncometabolites despite tumor metabolic heterogeneity.
Collapse
Affiliation(s)
- Chandrima Das
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, 700064, India.
- Homi Bhabha National Institute, Mumbai, 400094, India.
| | - Apoorva Bhattacharya
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, 700064, India
| | - Swagata Adhikari
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, 700064, India
- Homi Bhabha National Institute, Mumbai, 400094, India
| | - Atanu Mondal
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, 700064, India
- Homi Bhabha National Institute, Mumbai, 400094, India
| | - Payel Mondal
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, 700064, India
- Homi Bhabha National Institute, Mumbai, 400094, India
| | - Santanu Adhikary
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, 700064, India
- Structural Biology and Bioinformatics Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, 700032, India
| | - Siddhartha Roy
- Structural Biology and Bioinformatics Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, 700032, India
| | - Kenneth Ramos
- Center for Genomics and Precision Medicine, Texas A&M University, School of Medicine, Houston, TX, 77030, USA
| | - Kamlesh K Yadav
- Center for Genomics and Precision Medicine, Texas A&M University, School of Medicine, Houston, TX, 77030, USA
- School of Engineering Medicine, Texas A&M University, School of Medicine, Houston, TX, 77030, USA
| | - John A Tainer
- The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Tej K Pandita
- Center for Genomics and Precision Medicine, Texas A&M University, School of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
13
|
Filipe EC, Velayuthar S, Philp A, Nobis M, Latham SL, Parker AL, Murphy KJ, Wyllie K, Major GS, Contreras O, Mok ETY, Enriquez RF, McGowan S, Feher K, Quek L, Hancock SE, Yam M, Tran E, Setargew YFI, Skhinas JN, Chitty JL, Phimmachanh M, Han JZR, Cadell AL, Papanicolaou M, Mahmodi H, Kiedik B, Junankar S, Ross SE, Lam N, Coulson R, Yang J, Zaratzian A, Da Silva AM, Tayao M, Chin IL, Cazet A, Kansara M, Segara D, Parker A, Hoy AJ, Harvey RP, Bogdanovic O, Timpson P, Croucher DR, Lim E, Swarbrick A, Holst J, Turner N, Choi YS, Kabakova IV, Philp A, Cox TR. Tumor Biomechanics Alters Metastatic Dissemination of Triple Negative Breast Cancer via Rewiring Fatty Acid Metabolism. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307963. [PMID: 38602451 PMCID: PMC11186052 DOI: 10.1002/advs.202307963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 03/11/2024] [Indexed: 04/12/2024]
Abstract
In recent decades, the role of tumor biomechanics on cancer cell behavior at the primary site has been increasingly appreciated. However, the effect of primary tumor biomechanics on the latter stages of the metastatic cascade, such as metastatic seeding of secondary sites and outgrowth remains underappreciated. This work sought to address this in the context of triple negative breast cancer (TNBC), a cancer type known to aggressively disseminate at all stages of disease progression. Using mechanically tuneable model systems, mimicking the range of stiffness's typically found within breast tumors, it is found that, contrary to expectations, cancer cells exposed to softer microenvironments are more able to colonize secondary tissues. It is shown that heightened cell survival is driven by enhanced metabolism of fatty acids within TNBC cells exposed to softer microenvironments. It is demonstrated that uncoupling cellular mechanosensing through integrin β1 blocking antibody effectively causes stiff primed TNBC cells to behave like their soft counterparts, both in vitro and in vivo. This work is the first to show that softer tumor microenvironments may be contributing to changes in disease outcome by imprinting on TNBC cells a greater metabolic flexibility and conferring discrete cell survival advantages.
Collapse
|
14
|
Taunk K, Jajula S, Bhavsar PP, Choudhari M, Bhanuse S, Tamhankar A, Naiya T, Kalita B, Rapole S. The prowess of metabolomics in cancer research: current trends, challenges and future perspectives. Mol Cell Biochem 2024:10.1007/s11010-024-05041-w. [PMID: 38814423 DOI: 10.1007/s11010-024-05041-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: 12/21/2023] [Accepted: 05/18/2024] [Indexed: 05/31/2024]
Abstract
Cancer due to its heterogeneous nature and large prevalence has tremendous socioeconomic impacts on populations across the world. Therefore, it is crucial to discover effective panels of biomarkers for diagnosing cancer at an early stage. Cancer leads to alterations in cell growth and differentiation at the molecular level, some of which are very unique. Therefore, comprehending these alterations can aid in a better understanding of the disease pathology and identification of the biomolecules that can serve as effective biomarkers for cancer diagnosis. Metabolites, among other biomolecules of interest, play a key role in the pathophysiology of cancer whose levels are significantly altered while 'reprogramming the energy metabolism', a cellular condition favored in cancer cells which is one of the hallmarks of cancer. Metabolomics, an emerging omics technology has tremendous potential to contribute towards the goal of investigating cancer metabolites or the metabolic alterations during the development of cancer. Diverse metabolites can be screened in a variety of biofluids, and tumor tissues sampled from cancer patients against healthy controls to capture the altered metabolism. In this review, we provide an overview of different metabolomics approaches employed in cancer research and the potential of metabolites as biomarkers for cancer diagnosis. In addition, we discuss the challenges associated with metabolomics-driven cancer research and gaze upon the prospects of this emerging field.
Collapse
Affiliation(s)
- Khushman Taunk
- Proteomics Lab, National Centre for Cell Science, Ganeshkhind, Pune, Maharashtra, 411007, India
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, West Bengal, NH12 Simhat, Haringhata, Nadia, West Bengal, 741249, India
| | - Saikiran Jajula
- Proteomics Lab, National Centre for Cell Science, Ganeshkhind, Pune, Maharashtra, 411007, India
| | - Praneeta Pradip Bhavsar
- Proteomics Lab, National Centre for Cell Science, Ganeshkhind, Pune, Maharashtra, 411007, India
| | - Mahima Choudhari
- Proteomics Lab, National Centre for Cell Science, Ganeshkhind, Pune, Maharashtra, 411007, India
| | - Sadanand Bhanuse
- Proteomics Lab, National Centre for Cell Science, Ganeshkhind, Pune, Maharashtra, 411007, India
| | - Anup Tamhankar
- Department of Surgical Oncology, Deenanath Mangeshkar Hospital and Research Centre, Erandawne, Pune, Maharashtra, 411004, India
| | - Tufan Naiya
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, West Bengal, NH12 Simhat, Haringhata, Nadia, West Bengal, 741249, India
| | - Bhargab Kalita
- Proteomics Lab, National Centre for Cell Science, Ganeshkhind, Pune, Maharashtra, 411007, India.
- Amrita School of Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Ponekkara, Kochi, Kerala, 682041, India.
| | - Srikanth Rapole
- Proteomics Lab, National Centre for Cell Science, Ganeshkhind, Pune, Maharashtra, 411007, India.
| |
Collapse
|
15
|
Sunassee ED, Deutsch RJ, D’Agostino VW, Castellano-Escuder P, Siebeneck EA, Ilkayeva O, Crouch BT, Madonna MC, Everitt J, Alvarez JV, Palmer GM, Hirschey MD, Ramanujam N. Optical imaging reveals chemotherapy-induced metabolic reprogramming of residual disease and recurrence. SCIENCE ADVANCES 2024; 10:eadj7540. [PMID: 38579004 PMCID: PMC10997195 DOI: 10.1126/sciadv.adj7540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 03/04/2024] [Indexed: 04/07/2024]
Abstract
Fewer than 20% of triple-negative breast cancer patients experience long-term responses to mainstay chemotherapy. Resistant tumor subpopulations use alternative metabolic pathways to escape therapy, survive, and eventually recur. Here, we show in vivo, longitudinal metabolic reprogramming in residual disease and recurrence of triple-negative breast cancer xenografts with varying sensitivities to the chemotherapeutic drug paclitaxel. Optical imaging coupled with metabolomics reported an increase in non-glucose-driven mitochondrial metabolism and an increase in intratumoral metabolic heterogeneity during regression and residual disease in resistant MDA-MB-231 tumors. Conversely, sensitive HCC-1806 tumors were primarily reliant on glucose uptake and minimal changes in metabolism or heterogeneity were observed over the tumors' therapeutic life cycles. Further, day-matched resistant HCC-1806 tumors revealed a higher reliance on mitochondrial metabolism and elevated metabolic heterogeneity compared to sensitive HCC-1806 tumors. Together, metabolic flexibility, increased reliance on mitochondrial metabolism, and increased metabolic heterogeneity are defining characteristics of persistent residual disease, features that will inform the appropriate type and timing of therapies.
Collapse
Affiliation(s)
| | - Riley J. Deutsch
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | - Pol Castellano-Escuder
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Durham, NC, USA
- Department of Pharmacology and Cancer Biology, School of Medicine, Duke University, Durham, NC, USA
- Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University Medical Center, Durham, NC, USA
| | | | - Olga Ilkayeva
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Durham, NC, USA
- Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University Medical Center, Durham, NC, USA
| | - Brian T. Crouch
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Megan C. Madonna
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Jeffrey Everitt
- Department of Pathology, School of Medicine, Duke University, Durham, NC, USA
| | - James V. Alvarez
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Matthew D. Hirschey
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Durham, NC, USA
- Department of Pharmacology and Cancer Biology, School of Medicine, Duke University, Durham, NC, USA
- Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University Medical Center, Durham, NC, USA
| | - Nirmala Ramanujam
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- Department of Radiation Oncology, Duke University, Durham, NC, USA
| |
Collapse
|
16
|
Mitaishvili E, Feinsod H, David Z, Shpigel J, Fernandez C, Sauane M, de la Parra C. The Molecular Mechanisms behind Advanced Breast Cancer Metabolism: Warburg Effect, OXPHOS, and Calcium. FRONT BIOSCI-LANDMRK 2024; 29:99. [PMID: 38538285 PMCID: PMC10999756 DOI: 10.31083/j.fbl2903099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/24/2024] [Accepted: 02/22/2024] [Indexed: 04/10/2024]
Abstract
Altered metabolism represents a fundamental difference between cancer cells and normal cells. Cancer cells have a unique ability to reprogram their metabolism by deviating their reliance from primarily oxidative phosphorylation (OXPHOS) to glycolysis, in order to support their survival. This metabolic phenotype is referred to as the "Warburg effect" and is associated with an increase in glucose uptake, and a diversion of glycolytic intermediates to alternative pathways that support anabolic processes. These processes include synthesis of nucleic acids, lipids, and proteins, necessary for the rapidly dividing cancer cells, sustaining their growth, proliferation, and capacity for successful metastasis. Triple-negative breast cancer (TNBC) is one of the most aggressive subtypes of breast cancer, with the poorest patient outcome due to its high rate of metastasis. TNBC is characterized by elevated glycolysis and in certain instances, low OXPHOS. This metabolic dysregulation is linked to chemotherapeutic resistance in TNBC research models and patient samples. There is more than a single mechanism by which this metabolic switch occurs and here, we review the current knowledge of relevant molecular mechanisms involved in advanced breast cancer metabolism, focusing on TNBC. These mechanisms include the Warburg effect, glycolytic adaptations, microRNA regulation, mitochondrial involvement, mitochondrial calcium signaling, and a more recent player in metabolic regulation, JAK/STAT signaling. In addition, we explore some of the drugs and compounds targeting cancer metabolic reprogramming. Research on these mechanisms is highly promising and could ultimately offer new opportunities for the development of innovative therapies to treat advanced breast cancer characterized by dysregulated metabolism.
Collapse
Affiliation(s)
- Erna Mitaishvili
- Department of Chemistry, Herbert H. Lehman College, City University of New York, New York, NY 10468, USA
- PhD Program in Biology, The Graduate Center, City University of New York, New York, NY 10016, USA
| | - Hanna Feinsod
- Department of Chemistry, Herbert H. Lehman College, City University of New York, New York, NY 10468, USA
- Department of Chemistry, Columbia University, New York, NY 10027, USA
| | - Zachary David
- Department of Chemistry, Herbert H. Lehman College, City University of New York, New York, NY 10468, USA
| | - Jessica Shpigel
- Department of Chemistry, Herbert H. Lehman College, City University of New York, New York, NY 10468, USA
| | - Chelsea Fernandez
- Department of Chemistry, Herbert H. Lehman College, City University of New York, New York, NY 10468, USA
| | - Moira Sauane
- PhD Program in Biology, The Graduate Center, City University of New York, New York, NY 10016, USA
- Department of Biological Sciences, Herbert H. Lehman College, City University of New York, New York, NY 10468, USA
| | - Columba de la Parra
- Department of Chemistry, Herbert H. Lehman College, City University of New York, New York, NY 10468, USA
- PhD Program in Biology, The Graduate Center, City University of New York, New York, NY 10016, USA
- PhD Programs in Biochemistry and Chemistry, The Graduate Center, City University of New York, New York, NY 10016, USA
| |
Collapse
|
17
|
Singh P, Sen K, Sa P, Khuntia A, Raghav SK, Swain RK, Sahoo SK. Piperlongumine based nanomedicine impairs glycolytic metabolism in triple negative breast cancer stem cells through modulation of GAPDH & FBP1. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 123:155181. [PMID: 38091824 DOI: 10.1016/j.phymed.2023.155181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/27/2023] [Accepted: 11/01/2023] [Indexed: 01/17/2024]
Abstract
BACKGROUND Triple negative breast cancer (TNBC) is the most aggressive subtype of breast cancer and exhibits high rate of chemoresistance, metastasis, and relapse. This can be attributed to the failure of conventional therapeutics to target a sub-population of slow cycling or quiescent cells called as cancer stem cells (CSCs). Therefore, elimination of CSCs is essential for effective TNBC treatment. PURPOSE Research suggests that breast CSCs exhibit elevated glycolytic metabolism which directly contributes in maintenance of stemness, self-renewability and chemoresistance as well as in tumor progression. Therefore, this study aimed to target rewired metabolism which can serve as Achilles heel for CSCs population and have far reaching effect in TNBC treatment. METHODS We used two preclinical models, zebrafish and nude mice to evaluate the fate of nanoparticles as well as the therapeutic efficacy of both piperlongumine (PL) and its nanomedicine (PL-NPs). RESULTS In this context, we explored a phytochemical piperlongumine (PL) which has potent anti-cancer properties but poor pharmacokinetics impedes its clinical translation. So, we developed PLGA based nanomedicine for PL (PL-NPs), and demonstrated that it overcomes the pharmacokinetic limitations of PL, along with imparting advantages of selective tumor targeting through Enhanced Permeability and Retention (EPR) effect in zebrafish xenograft model. Further, we demonstrated that PL-NPs efficiently inhibit glycolysis in CSCs through inhibition of glyceraldehyde-3-phosphate dehydrogenase (GAPDH) by modulating glutathione S-transferase pi 1 (GSTP1) and upregulation of fructose-1,6-bisphosphatase 1 (FBP1), a rate-limiting enzyme in gluconeogenesis. We also illustrated that inhibition of glycolysis results in overall tumor regression in two preclinical models. CONCLUSION This study discusses novel mechanism of action by which PL acts on CSCSs. Taken together our study provides insight into development of PL based nanomedicine which could be exploited in clinics to achieve complete eradication of TNBC by targeting CSCs.
Collapse
Affiliation(s)
- Priya Singh
- Institute of Life Sciences, Nalco square, Bhubaneswar 751 023, Odisha, India; Regional Centre for Biotechnology, Faridabad-Gurgaon Expressway, Faridabad 121 001, Haryana, India
| | - Kaushik Sen
- Institute of Life Sciences, Nalco square, Bhubaneswar 751 023, Odisha, India; Regional Centre for Biotechnology, Faridabad-Gurgaon Expressway, Faridabad 121 001, Haryana, India
| | - Pratikshya Sa
- Institute of Life Sciences, Nalco square, Bhubaneswar 751 023, Odisha, India; Regional Centre for Biotechnology, Faridabad-Gurgaon Expressway, Faridabad 121 001, Haryana, India
| | - Auromira Khuntia
- Institute of Life Sciences, Nalco square, Bhubaneswar 751 023, Odisha, India; Regional Centre for Biotechnology, Faridabad-Gurgaon Expressway, Faridabad 121 001, Haryana, India
| | - Sunil K Raghav
- Institute of Life Sciences, Nalco square, Bhubaneswar 751 023, Odisha, India
| | - Rajeeb K Swain
- Institute of Life Sciences, Nalco square, Bhubaneswar 751 023, Odisha, India
| | - Sanjeeb Kumar Sahoo
- Institute of Life Sciences, Nalco square, Bhubaneswar 751 023, Odisha, India.
| |
Collapse
|
18
|
Yamashita N, Withers H, Morimoto Y, Bhattacharya A, Haratake N, Daimon T, Fushimi A, Nakashoji A, Thorner AR, Isenhart E, Rosario S, Long MD, Kufe D. MUC1-C integrates aerobic glycolysis with suppression of oxidative phosphorylation in triple-negative breast cancer stem cells. iScience 2023; 26:108168. [PMID: 37915591 PMCID: PMC10616323 DOI: 10.1016/j.isci.2023.108168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/17/2023] [Accepted: 10/05/2023] [Indexed: 11/03/2023] Open
Abstract
Activation of the MUC1-C protein promotes lineage plasticity, epigenetic reprogramming, and the cancer stem cell (CSC) state. The present studies performed on enriched populations of triple-negative breast cancer (TNBC) CSCs demonstrate that MUC1-C is essential for integrating activation of glycolytic pathway genes with self-renewal and tumorigenicity. MUC1-C further integrates the glycolytic pathway with suppression of mitochondrial DNA (mtDNA) genes encoding components of mitochondrial Complexes I-V. The repression of mtDNA genes is explained by MUC1-C-mediated (i) downregulation of the mitochondrial transcription factor A (TFAM) required for mtDNA transcription and (ii) induction of the mitochondrial transcription termination factor 3 (mTERF3). In support of pathogenesis that suppresses mitochondrial ROS production, targeting MUC1-C increases (i) mtDNA gene transcription, (ii) superoxide levels, and (iii) loss of self-renewal capacity. These findings and scRNA-seq analysis of CSC subpopulations indicate that MUC1-C regulates self-renewal and redox balance by integrating activation of glycolysis with suppression of oxidative phosphorylation.
Collapse
Affiliation(s)
- Nami Yamashita
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Henry Withers
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | | | | | - Naoki Haratake
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Tatsuaki Daimon
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Atsushi Fushimi
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Ayako Nakashoji
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Aaron R. Thorner
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Emily Isenhart
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Spencer Rosario
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Mark D. Long
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Donald Kufe
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
19
|
Gallo M, Ferrari E, Terrazzan A, Brugnoli F, Spisni A, Taccioli C, Aguiari G, Trentini A, Volinia S, Keillor JW, Bergamini CM, Bianchi N, Pertinhez TA. Metabolic characterisation of transglutaminase 2 inhibitor effects in breast cancer cell lines. FEBS J 2023; 290:5411-5433. [PMID: 37597264 DOI: 10.1111/febs.16931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 07/04/2023] [Accepted: 08/09/2023] [Indexed: 08/21/2023]
Abstract
Transglutaminase 2 (TG2), which mediates post-translational modifications of multiple intracellular enzymes, is involved in the pathogenesis and progression of cancer. We used 1 H-NMR metabolomics to study the effects of AA9, a novel TG2 inhibitor, on two breast cancer cell lines with distinct phenotypes, MCF-7 and MDA-MB-231. AA9 can promote apoptosis in both cell lines, but it is particularly effective in MD-MB-231, inhibiting transamidation reactions and decreasing cell migration and invasiveness. This metabolomics study provides evidence of a major effect of AA9 on MDA-MB-231 cells, impacting glutamate and aspartate metabolism, rather than on MCF-7 cells, characterised by choline and O-phosphocholine decrease. Interestingly, AA9 treatment induces myo-inositol alteration in both cell lines, indicating action on phosphatidylinositol metabolism, likely modulated by the G protein activity of TG2 on phospholipase C. Considering the metabolic deregulations that characterise various breast cancer subtypes, the existence of a metabolic pathway affected by AA9 further points to TG2 as a promising hot spot. The metabolomics approach provides a powerful tool to monitor the effectiveness of inhibitors and better understand the role of TG2 in cancer.
Collapse
Affiliation(s)
- Mariana Gallo
- Department of Medicine and Surgery, University of Parma, Italy
| | - Elena Ferrari
- Department of Medicine and Surgery, University of Parma, Italy
| | - Anna Terrazzan
- Department of Translational Medicine, University of Ferrara, Italy
| | | | - Alberto Spisni
- Department of Medicine and Surgery, University of Parma, Italy
| | - Cristian Taccioli
- Department of Animal Medicine, Production and Health (MAPS), University of Padua, Italy
| | - Gianluca Aguiari
- Department of Neuroscience and Rehabilitation, University of Ferrara, Italy
| | - Alessandro Trentini
- Department of Environmental Sciences and Prevention, University of Ferrara, Italy
| | - Stefano Volinia
- Department of Translational Medicine, University of Ferrara, Italy
| | - Jeffrey W Keillor
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Canada
| | - Carlo M Bergamini
- Department of Neuroscience and Rehabilitation, University of Ferrara, Italy
| | | | | |
Collapse
|
20
|
Granit Mizrahi A, Gugenheim A, Hamad H, Hamed R, Tetro N, Maimon O, Khutsurauli S, Nechushtan H, Nisman B, Duran D, Samman W, Birimberg-Schwartz L, Grunewald M, Eyal S, Peretz T. Valproic acid reprograms the metabolic aberration of cisplatin treatment via ALDH modulation in triple-negative breast cancer cells. Front Cell Dev Biol 2023; 11:1217149. [PMID: 37954205 PMCID: PMC10639136 DOI: 10.3389/fcell.2023.1217149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 10/02/2023] [Indexed: 11/14/2023] Open
Abstract
We recently demonstrated that the histone deacetylase inhibitor valproic acid (VPA) reprograms the cisplatin-induced metabolome of triple-negative breast cancer (TNBC) cells, including a shift in hexose levels. Accordingly, here, we tested the hypothesis that VPA alters glucose metabolism in correlation with cisplatin sensitivity. Two TNBC cell lines, MDA-MB-231 (a cisplatin-resistant line) and MDA-MB-436 (a cisplatin-sensitive line), were analyzed. The glycolysis and oxidative metabolism were measured using the Glycolysis Stress Test kit. The expression of aldehyde dehydrogenases (ALDHs), enzymes linked to drug resistance, was investigated by Western blot and real-time PCR analyses. We additionally studied the influence of ALDH inhibition by disulfiram on the viability of MDA-MB-231 cells and on a TNBC patient-derived organoid system. Cisplatin treatment reduced the extracellular acidification rate in MDA-MB-436 cells but not MDA-MB-231 cells, whereas VPA addition increased the extracellular acidification rate in both cell lines. VPA further reduced the oxygen consumption rate of cisplatin-treated MDA-MB-436 cells, which correlated with cell cycle alterations. However, in MDA-MB-231 cells, the cell cycle distribution did not change between cisplatin/VPA-cisplatin treatments. In both cell lines, VPA increased the expression of ALDH isoform and ALDH1A1 expression. However, only in MDA-MB-231 cells, VPA synergized with cisplatin to augment this effect. Disulfiram sensitized the cells to the cytotoxic effects of the VPA-cisplatin combination. Furthermore, the disulfiram-VPA-chemotherapy combination was most effective in TNBC organoids. Our results show that ALDH overexpression may act as one mechanism of cellular resistance to VPA in TNBC and that its inhibition may enhance the therapeutic efficacy of VPA-chemotherapeutic drug combinations.
Collapse
Affiliation(s)
- Avital Granit Mizrahi
- Oncology Laboratory, Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
- School of Pharmacy, Institute for Drug Research, The Hebrew University, Jerusalem, Israel
| | - Ahinoam Gugenheim
- Oncology Laboratory, Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Haneen Hamad
- Oncology Laboratory, Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Roa’a Hamed
- School of Pharmacy, Institute for Drug Research, The Hebrew University, Jerusalem, Israel
| | - Nino Tetro
- School of Pharmacy, Institute for Drug Research, The Hebrew University, Jerusalem, Israel
| | - Ofra Maimon
- Oncology Laboratory, Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Salome Khutsurauli
- Oncology Laboratory, Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Hovav Nechushtan
- Oncology Laboratory, Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Benjamin Nisman
- Oncology Laboratory, Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Deborah Duran
- Faculty of Medicine, Hebrew University, Jerusalem, Israel
- Hadassah Organoid Center, The Hadassah Medical Organization, Jerusalem, Israel
| | - Widad Samman
- Faculty of Medicine, Hebrew University, Jerusalem, Israel
- Hadassah Organoid Center, The Hadassah Medical Organization, Jerusalem, Israel
| | - Liron Birimberg-Schwartz
- Faculty of Medicine, Hebrew University, Jerusalem, Israel
- Hadassah Organoid Center, The Hadassah Medical Organization, Jerusalem, Israel
- Department of Pediatric Gastroenterology, The Hadassah Medical Organization, Jerusalem, Israel
| | - Myriam Grunewald
- Faculty of Medicine, Hebrew University, Jerusalem, Israel
- Hadassah Organoid Center, The Hadassah Medical Organization, Jerusalem, Israel
| | - Sara Eyal
- School of Pharmacy, Institute for Drug Research, The Hebrew University, Jerusalem, Israel
| | - Tamar Peretz
- Oncology Laboratory, Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
- Faculty of Medicine, Hebrew University, Jerusalem, Israel
| |
Collapse
|
21
|
Zhang H, Gao A, Liu Q, Zhang F, Wang S, Chen X, Shi W, Zhang Y, Liu Q, Zheng Y, Sun Y. ILT4 reprograms glucose metabolism to promote tumor progression in triple-negative breast cancer. J Cell Sci 2023; 136:jcs260964. [PMID: 37622462 DOI: 10.1242/jcs.260964] [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: 01/15/2023] [Accepted: 08/16/2023] [Indexed: 08/26/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive and poorly treated subtype of breast cancer. Identifying novel drivers and mechanisms for tumor progression is essential for precise targeted therapy of TNBC. Immunoglobulin-like transcript 4 (ILT4; also known as LILRB2) is a classic myeloid suppressor for their activation and immune response. Our recent results found that ILT4 is also highly expressed in lung cancer cells, where it has a role in promoting immune evasion and thus tumor formation. However, the expression and function of ILT4 in breast cancer remains elusive. Here, using our patient cohort and public database analysis, we found that TNBC displayed the most abundant ILT4 expression among all breast cancer subtypes. Functionally, enriched ILT4 promoted TNBC cell proliferation, migration and invasion in vitro, as well as tumor growth and metastasis in vivo. Further mechanistic analysis revealed that ILT4 reprogrammed aerobic glycolysis of tumor cells via AKT-mTOR signaling-mediated glucose transporter 3 (GLUT3; also known as SLC2A3) and pyruvate kinase muscle 2 (PKM2, an isoform encoded by PKM) overexpression. ILT4 inhibition in TNBC reduced tumor progression and GLUT3 and PKM2 expression in vivo. Our study identified a novel driver for TNBC progression and proposed a promising strategy to combat TNBC by targeting ILT4.
Collapse
Affiliation(s)
- Haiqin Zhang
- Department of Oncology, Jinan Central Hospital, Shandong University, Jinan, 250013 Shandong, P. R. China
- Department of Oncology, Central hospital affiliated to Shandong First Medical University, Jinan, 250013 Shandong, P. R. China
- Research Center of Translational Medicine, Laboratory Animal Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013 Shandong, P. R. China
| | - Aiqin Gao
- Department of Thoracic Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong Academy of Medical Sciences, Shandong First Medical University, Jinan, 250117 Shandong, P. R. China
| | - Qiaohong Liu
- Department of Ultrasound, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013 Shandong, P. R. China
| | - Fang Zhang
- Department of Oncology, Central hospital affiliated to Shandong First Medical University, Jinan, 250013 Shandong, P. R. China
- Research Center of Translational Medicine, Laboratory Animal Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013 Shandong, P. R. China
| | - Shuyun Wang
- Phase I Clinical Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117 Shandong, P. R. China
| | - Xiaozheng Chen
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117 Shandong, P. R. China
| | - Wenjing Shi
- Jinan Central Hospital, Shandong University, Jinan, 250013 Shandong, P. R. China
| | - Ye Zhang
- Department of Oncology, Jinan Central Hospital, Weifang Medical University, Weifang, 250013 Shandong, P. R. China
| | - Qian Liu
- Department of Oncology, Jinan Central Hospital, Weifang Medical University, Weifang, 250013 Shandong, P. R. China
| | - Yan Zheng
- Research Center of Translational Medicine, Laboratory Animal Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013 Shandong, P. R. China
| | - Yuping Sun
- Phase I Clinical Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117 Shandong, P. R. China
- Phase I Clinical Research Center, Shandong University Cancer Center, Shandong Cancer Hospital and Institute, Jinan, 250117 Shandong, P. R. China
| |
Collapse
|
22
|
Sun R, Ge W, Zhu Y, Sayad A, Luna A, Lyu M, Liang S, Tobalina L, Rajapakse VN, Yu C, Zhang H, Fang J, Wu F, Xie H, Saez-Rodriguez J, Ying H, Reinhold WC, Sander C, Pommier Y, Neel BG, Aebersold R, Guo T. Proteomic Dynamics of Breast Cancer Cell Lines Identifies Potential Therapeutic Protein Targets. Mol Cell Proteomics 2023; 22:100602. [PMID: 37343696 PMCID: PMC10392136 DOI: 10.1016/j.mcpro.2023.100602] [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: 08/24/2022] [Revised: 04/18/2023] [Accepted: 06/12/2023] [Indexed: 06/23/2023] Open
Abstract
Treatment and relevant targets for breast cancer (BC) remain limited, especially for triple-negative BC (TNBC). We identified 6091 proteins of 76 human BC cell lines using data-independent acquisition (DIA). Integrating our proteomic findings with prior multi-omics datasets, we found that including proteomics data improved drug sensitivity predictions and provided insights into the mechanisms of action. We subsequently profiled the proteomic changes in nine cell lines (five TNBC and four non-TNBC) treated with EGFR/AKT/mTOR inhibitors. In TNBC, metabolism pathways were dysregulated after EGFR/mTOR inhibitor treatment, while RNA modification and cell cycle pathways were affected by AKT inhibitor. This systematic multi-omics and in-depth analysis of the proteome of BC cells can help prioritize potential therapeutic targets and provide insights into adaptive resistance in TNBC.
Collapse
Affiliation(s)
- Rui Sun
- Westlake Intelligent Biomarker Discovery Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Weigang Ge
- Bioinformatics Department, Westlake Omics (Hangzhou) Biotechnology Co, Ltd, Hangzhou, Zhejiang, China
| | - Yi Zhu
- Westlake Intelligent Biomarker Discovery Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China; Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Azin Sayad
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada; Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, New York, USA
| | - Augustin Luna
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA; Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Mengge Lyu
- Westlake Intelligent Biomarker Discovery Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Shuang Liang
- Westlake Intelligent Biomarker Discovery Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Luis Tobalina
- Bioinformatics and Data Science, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Vinodh N Rajapakse
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Chenhuan Yu
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang, China
| | - Huanhuan Zhang
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang, China
| | - Jie Fang
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang, China
| | - Fang Wu
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang, China
| | - Hui Xie
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang, China
| | - Julio Saez-Rodriguez
- Faculty of Medicine, Institute for Computational Biomedicine, Heidelberg University Hospital, BioQuant, Heidelberg University, Heidelberg, Baden-Württemberg, Germany
| | - Huazhong Ying
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - William C Reinhold
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Chris Sander
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA; Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Yves Pommier
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Benjamin G Neel
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada; Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, New York, USA.
| | - Ruedi Aebersold
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland; Faculty of Science, University of Zurich, Zurich, Switzerland.
| | - Tiannan Guo
- Westlake Intelligent Biomarker Discovery Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China; Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
23
|
Arai N, Hattori N, Yamashita S, Liu YY, Ebata T, Takeuchi C, Takeshima H, Fujii S, Kondo H, Mukai H, Ushijima T. HSD17B4 methylation enhances glucose dependence of BT-474 breast cancer cells and increases lapatinib sensitivity. Breast Cancer Res Treat 2023:10.1007/s10549-023-07013-y. [PMID: 37378696 DOI: 10.1007/s10549-023-07013-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023]
Abstract
PURPOSE HER2-positive breast cancer has a high chance of achieving pathological complete response when HSD17B4, responsible for peroxisomal β-oxidation of very long-chain fatty acids (VLCFA) and estradiol, is methylation-silenced. Here, we aimed to identify the underlying molecular mechanism. METHODS Using a HER2-positive breast cancer cell line, BT-474, control and knock-out (KO) clones were obtained. Metabolic characteristics were analyzed using a Seahorse Flux analyzer. RESULTS HSD17B4 KO suppressed cellular proliferation, and enhanced sensitivity to lapatinib approximately tenfold. The KO led to accumulation of VLCFA and a decrease of polyunsaturated fatty acids (PUFAs), such as docosahexaenoic acid (DHA) and arachidonic acid. HSD17B4 KO increased Akt phosphorylation, possibly via decreased DHA, and genes involved in oxidative phosphorylation (OxPhos) and electron transport chain (ETC) were upregulated. Increased mitochondrial ATP production in the KO cells was confirmed by extracellular flux analyzer. Increased OxPhos led to severe dependence of the KO cells on pyruvate from glycolysis. Suppression of glycolysis by lapatinib led to severe delayed suppression of OxPhos in KO cells. CONCLUSION HSD17B4 KO in BT-474 cells caused a decrease of PUFAs, increased Akt phosphorylation, enhanced glucose dependence of OxPhos, and increased sensitivity to inhibition of HER2, upstream of Akt. This mechanism may be applicable to other HER2-positive glucose-dependent breast cancer cells with HSD17B4 silencing.
Collapse
Affiliation(s)
- Nobuaki Arai
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan
- Department of Surgery, Kyorin University School of Medicine, Tokyo, Japan
| | - Naoko Hattori
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan
- Department of Epigenomics, Institute for Advanced Life Sciences, Hoshi University, Tokyo, Japan
| | - Satoshi Yamashita
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Yu-Yu Liu
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan
- Department of Epigenomics, Institute for Advanced Life Sciences, Hoshi University, Tokyo, Japan
| | - Takahiro Ebata
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan
- Department of Epigenomics, Institute for Advanced Life Sciences, Hoshi University, Tokyo, Japan
| | - Chihiro Takeuchi
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan
- Department of Epigenomics, Institute for Advanced Life Sciences, Hoshi University, Tokyo, Japan
| | - Hideyuki Takeshima
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan
- Department of Epigenomics, Institute for Advanced Life Sciences, Hoshi University, Tokyo, Japan
| | - Satoshi Fujii
- Department of Molecular Pathology, Yokohama City University Graduate School of Medicine, Kanagawa, Japan
| | - Haruhiko Kondo
- Department of Surgery, Kyorin University School of Medicine, Tokyo, Japan
| | - Hirofumi Mukai
- Department of Medical Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Toshikazu Ushijima
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan.
- Department of Epigenomics, Institute for Advanced Life Sciences, Hoshi University, Tokyo, Japan.
| |
Collapse
|
24
|
Zhang X, Sarkar S, Ashokan A, Surnar B, Kolishetti N, Dhar S. All-in-One Pyruvate Dehydrogenase Kinase Inhibitor for Tracking, Targeting, and Enhanced Efficacy. Bioconjug Chem 2023; 34:1122-1129. [PMID: 37279374 PMCID: PMC10903362 DOI: 10.1021/acs.bioconjchem.3c00152] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The metabolic preference of cells toward glycolysis often indicates a diseased state ranging from cancer to other dysfunctions. When a particular cell type utilizes glycolysis as a major energy production pathway, their mitochondria become impaired resulting a cascade of events which eventually contributes to resistance toward therapies to tackle such diseases. In abnormal tissues such as seen in the tumor microenvironment, when cancer cells utilize glycolysis, other cell types such as the immune cells switch their metabolism and prefer such glycolysis. As a result, utilization of therapies to destroy glycolytic preferences by cancer cells results in destruction of immune cells contributing toward an immunosuppressive phenotype. Thus, development of targeted, trackable, comparatively stable glycolysis inhibitors is urgently needed to manage diseases where glycolysis is preferred for disease progression. No glycolysis inhibitor exists which can be tracked and packaged in a delivery vehicle for efficient targeted deployment. Here, we report synthesis, characterization, and formulation of an all-in-one glycolysis inhibitor and document the therapeutic potential along with trackability and glycolysis inhibition of this inhibitor by utilizing an in vivo breast cancer model.
Collapse
Affiliation(s)
- Xiao Zhang
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, University of Miami, Miami, Florida 33136, United States
- Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, University of Miami, Miami, Florida 33136, United States
| | - Shrita Sarkar
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, University of Miami, Miami, Florida 33136, United States
- Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, University of Miami, Miami, Florida 33136, United States
| | - Akash Ashokan
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, University of Miami, Miami, Florida 33136, United States
- Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, University of Miami, Miami, Florida 33136, United States
| | - Bapurao Surnar
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, University of Miami, Miami, Florida 33136, United States
- Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, University of Miami, Miami, Florida 33136, United States
| | - Nagesh Kolishetti
- Department of Immunology and Nano-Medicine, Herbert Wertheim, College of Medicine, Florida International University, Miami, Florida 33199, United States
- Institute of Neuroimmune Pharmacology, Herbert Wertheim College of Medicine, Miami, Florida 33199, United States
| | - Shanta Dhar
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, University of Miami, Miami, Florida 33136, United States
- Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, University of Miami, Miami, Florida 33136, United States
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida 33146, United States
| |
Collapse
|
25
|
Rajoria B, Zhang X, Yee D. IGF-1 Stimulates Glycolytic ATP Production in MCF-7L Cells. Int J Mol Sci 2023; 24:10209. [PMID: 37373357 PMCID: PMC10299323 DOI: 10.3390/ijms241210209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/08/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
The Insulin-like Growth Factor (IGF) system in breast cancer progression has been a matter of interest for decades, but targeting this system did not result in a successful clinical strategy. The system's complexity and homology of its two receptors-insulin receptor (IR) and type 1 insulin-like growth factor receptor (IGF-1R)-are possible causes. The IGF system maintains cell proliferation and also regulates metabolism, making it a pathway to explore. To understand the metabolic phenotype of breast cancer cells, we quantified their real-time ATP production rate upon acute stimulation with ligands-insulin-like growth factor 1 (1GF-1) and insulin. MCF-7L cells express both IGF-1R and IR, while tamoxifen-resistant MCF-7L (MCF-7L TamR) cells have downregulated IGF-1R with unchanged IR levels. Treating MCF-7L cells with 5 nM IGF-1 increased the glycolytic ATP production rate, while 10 nM insulin did not affect metabolism when compared with the control. Neither treatment altered ATP production in MCF-7L TamR cells. This study provides evidence of the relationship between metabolic dysfunction, cancer, and the IGF axis. In these cells, IGF-1R, and not IR, regulates ATP production.
Collapse
Affiliation(s)
- Bhumika Rajoria
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA;
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Xihong Zhang
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Douglas Yee
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA;
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA;
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
26
|
Mussa A, Afolabi HA, Syed NH, Talib M, Murtadha AH, Hajissa K, Mokhtar NF, Mohamud R, Hassan R. The NF-κB Transcriptional Network Is a High-Dose Vitamin C-Targetable Vulnerability in Breast Cancer. Biomedicines 2023; 11:biomedicines11041060. [PMID: 37189677 DOI: 10.3390/biomedicines11041060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/24/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
Breast cancer (BC) is the most common cancer type among women with a distinct clinical presentation, but the survival rate remains moderate despite advances in multimodal therapy. Consequently, a deeper understanding of the molecular etiology is required for the development of more effective treatments for BC. The relationship between inflammation and tumorigenesis is well established, and the activation of the pro-inflammatory transcription factor nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) is frequently identified in BC. Constitutive NF-κB activation is linked to cell survival, metastasis, proliferation, and hormonal, chemo-, and radiotherapy resistance. Moreover, the crosstalk between NF-κB and other transcription factors is well documented. It is reported that vitamin C plays a key role in preventing and treating a number of pathological conditions, including cancer, when administered at remarkably high doses. Indeed, vitamin C can regulate the activation of NF-κB by inhibiting specific NF-κB-dependent genes and multiple stimuli. In this review, we examine the various NF-κB impacts on BC development. We also provide some insight into how the NF-κB network may be targeted as a potential vulnerability by using natural pro-oxidant therapies such as vitamin C.
Collapse
|
27
|
Jekabsons MB, Merrell M, Skubiz AG, Thornton N, Milasta S, Green D, Chen T, Wang YH, Avula B, Khan IA, Zhou YD. Breast cancer cells that preferentially metastasize to lung or bone are more glycolytic, synthesize serine at greater rates, and consume less ATP and NADPH than parent MDA-MB-231 cells. Cancer Metab 2023; 11:4. [PMID: 36805760 PMCID: PMC9940388 DOI: 10.1186/s40170-023-00303-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 01/25/2023] [Indexed: 02/22/2023] Open
Abstract
Gene expression signatures associated with breast cancer metastases suggest that metabolic re-wiring is important for metastatic growth in lungs, bones, and other organs. However, since pathway fluxes depend on additional factors such as ATP demand, allosteric effects, and post-translational modification, flux analysis is necessary to conclusively establish phenotypes. In this study, the metabolic phenotypes of breast cancer cell lines with low (T47D) or high (MDA-MB-231) metastatic potential, as well as lung (LM)- and bone (BoM)-homing lines derived from MDA-MB-231 cells, were assessed by 13C metabolite labeling from [1,2-13C] glucose or [5-13C] glutamine and the rates of nutrient and oxygen consumption and lactate production. MDA-MB-231 and T47D cells produced 55 and 63%, respectively, of ATP from oxidative phosphorylation, whereas LM and BoM cells were more glycolytic, deriving only 20-25% of their ATP from mitochondria. ATP demand by BoM and LM cells was approximately half the rate of the parent cells. Of the anabolic fluxes assessed, nucleotide synthesis was the major ATP consumer for all cell lines. Glycolytic NADH production by LM cells exceeded the rate at which it could be oxidized by mitochondria, suggesting that the malate-aspartate shuttle was not involved in re-oxidation of these reducing equivalents. Serine synthesis was undetectable in MDA-MB-231 cells, whereas 3-5% of glucose was shunted to serine by LM and BoM lines. Proliferation rates of T47D, BoM, and LM lines tightly correlated with their respiration-normalized NADPH production rates. In contrast, MDA-MB-231 cells produced NADPH and GSH at higher rates, suggesting this line is more oxidatively stressed. Approximately half to two-thirds of NADPH produced by T47D, MDA-MB-231, and BoM cells was from the oxidative PPP, whereas the majority in LM cells was from the folate cycle. All four cell lines used the non-oxidative PPP to produce pentose phosphates, although this was most prominent for LM cells. Taken together, the metabolic phenotypes of LM and BoM lines differed from the parent line and from each other, supporting the metabolic re-wiring hypothesis as a feature of metastasis to lung and bone.
Collapse
Affiliation(s)
- Mika B. Jekabsons
- grid.251313.70000 0001 2169 2489Department of Biology, University of Mississippi, University, MS 38677 USA
| | - Mollie Merrell
- grid.251313.70000 0001 2169 2489Department of Biology, University of Mississippi, University, MS 38677 USA
| | - Anna G. Skubiz
- grid.251313.70000 0001 2169 2489Department of Biology, University of Mississippi, University, MS 38677 USA
| | - Noah Thornton
- grid.251313.70000 0001 2169 2489Department of Biology, University of Mississippi, University, MS 38677 USA
| | - Sandra Milasta
- grid.240871.80000 0001 0224 711XDepartment of Immunology, St Jude Children’s Research Hospital, Memphis, TN 38105 USA
| | - Douglas Green
- grid.240871.80000 0001 0224 711XDepartment of Immunology, St Jude Children’s Research Hospital, Memphis, TN 38105 USA
| | - Taosheng Chen
- grid.240871.80000 0001 0224 711XDepartment of Chemical Biology and Therapeutics, St Jude Children’s Research Hospital, Memphis, TN 38105 USA
| | - Yan-Hong Wang
- grid.251313.70000 0001 2169 2489National Center for Natural Products Research, School of Pharmacy, University of Mississippi, University, MS 38677 USA
| | - Bharathi Avula
- grid.251313.70000 0001 2169 2489National Center for Natural Products Research, School of Pharmacy, University of Mississippi, University, MS 38677 USA
| | - Ikhlas A. Khan
- grid.251313.70000 0001 2169 2489National Center for Natural Products Research, School of Pharmacy, University of Mississippi, University, MS 38677 USA ,grid.251313.70000 0001 2169 2489Department of Biomedical Sciences, School of Pharmacy, University of Mississippi, University, MS 38677 USA
| | - Yu-Dong Zhou
- grid.251313.70000 0001 2169 2489Department of Chemistry and Biochemistry, University of Mississippi, University, MS 38677 USA
| |
Collapse
|
28
|
Chacon-Barahona JA, MacKeigan JP, Lanning NJ. Unique Metabolic Contexts Sensitize Cancer Cells and Discriminate between Glycolytic Tumor Types. Cancers (Basel) 2023; 15:cancers15041158. [PMID: 36831501 PMCID: PMC9953999 DOI: 10.3390/cancers15041158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/06/2023] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
Cancer cells utilize variable metabolic programs in order to maintain homeostasis in response to environmental challenges. To interrogate cancer cell reliance on glycolytic programs under different nutrient availabilities, we analyzed a gene panel containing all glycolytic genes as well as pathways associated with glycolysis. Using this gene panel, we analyzed the impact of an siRNA library on cellular viability in cells containing only glucose or only pyruvate as the major bioenergetic nutrient source. From these panels, we aimed to identify genes that elicited conserved and glycolysis-dependent changes in cellular bioenergetics across glycolysis-promoting and OXPHOS-promoting conditions. To further characterize gene sets within this panel and identify similarities and differences amongst glycolytic tumor RNA-seq profiles across a pan-cancer cohort, we then used unsupervised statistical classification of RNA-seq profiles for glycolytic cancers and non-glycolytic cancer types. Here, Kidney renal clear cell carcinoma (KIRC); Head and Neck squamous cell carcinoma (HNSC); and Lung squamous cell carcinoma (LUSC) defined the glycolytic cancer group, while Prostate adenocarcinoma (PRAD), Thyroid carcinoma (THCA), and Thymoma (THYM) defined the non-glycolytic cancer group. These groups were defined based on glycolysis scoring from previous studies, where KIRC, HNSC, and LUSC had the highest glycolysis scores, meanwhile, PRAD, THCA, and THYM had the lowest. Collectively, these results aimed to identify multi-omic profiles across cancer types with demonstrated variably glycolytic rates. Our analyses provide further support for strategies aiming to classify tumors by metabolic phenotypes in order to therapeutically target tumor-specific vulnerabilities.
Collapse
Affiliation(s)
| | - Jeffrey P. MacKeigan
- Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
- Department of Cell Biology, Van Andel Research Institute, Grand Rapids, MI 49503, USA
- Correspondence: (J.P.M.); (N.J.L.)
| | - Nathan J. Lanning
- Department of Biological Sciences, California State University, Los Angeles, CA 90032, USA
- Correspondence: (J.P.M.); (N.J.L.)
| |
Collapse
|
29
|
Li J, Gao R, Zhang J. USP22 Contributes to Chemoresistance, Stemness, and EMT Phenotype of Triple-Negative Breast Cancer Cells by egulating the Warburg Effect via c-Myc Deubiquitination. Clin Breast Cancer 2023; 23:162-175. [PMID: 36528490 DOI: 10.1016/j.clbc.2022.11.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 11/07/2022] [Accepted: 11/22/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Ubiquitin-specific protease 22 (USP22) has been implicated in the progression of breast cancer, while its regulatory functions in triple-negative breast cancer (TNBC) have been rarely reported. This study aimed to elucidate the effect and mechanism of USP22 on the malignant phenotype of TNBC cells. MATERIALS AND METHODS The expression of USP22, stemness genes, and EMT-related markers were analyzed by RT-qPCR and/or Western blotting. Cell stemness was determined by cell spheroid formation, flow cytometry for CD44+/CD24-, and extreme limiting dilution analysis. Cell proliferation and cisplatin (DDP) chemoresistance of TNBC cells were assessed by CCK-8 assay and xenograft model. Glycolysis was measured by Seahorse assay. The mechanism underlying the role of USP22 was explored by Co-immunoprecipitation, ubiquitination assay, and cycloheximide-chase analysis. RESULTS USP22 expression was positively correlated with DDP resistance in TNBC patients and cells. The proliferation, spheroid number, CD44+/CD24- cells, the expression of stemness genes and EMT-related markers in TNBC cells were significantly elevated after USP22 was overexpressed; however, these parameters in DDP-resistant TNBC (TNBC/DDP) cells were significantly reduced after silencing USP22. USP22 overexpression enhanced the extracellular acidification rate, proliferation, spheroid number, CD44+/CD24- cell number, and the expression of stemness genes and EMT-related markers in TNBC/DDP cells, while these effects were restrained by glycolysis inhibitors. Mechanically, USP22 interacted with c-Myc to promote its stabilization by deubiquitination in TNBC cells. Silencing of USP22 increased DDP sensitivity and survival of mice bearing TNBC. CONCLUSION USP22 contributes to chemoresistance, stemness, and EMT phenotype of TNBC cells by suppressing the glycolysis via c-Myc deubiquitination.
Collapse
Affiliation(s)
- Jie Li
- Department of General Surgery, Shanxi Provincial People's Hospital, Taiyuan, China.
| | - Runfang Gao
- Department of General Surgery, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Jing Zhang
- Department of General Surgery, Shanxi Provincial People's Hospital, Taiyuan, China
| |
Collapse
|
30
|
Schreier A, Zappasodi R, Serganova I, Brown KA, Demaria S, Andreopoulou E. Facts and Perspectives: Implications of tumor glycolysis on immunotherapy response in triple negative breast cancer. Front Oncol 2023; 12:1061789. [PMID: 36703796 PMCID: PMC9872136 DOI: 10.3389/fonc.2022.1061789] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 11/17/2022] [Indexed: 01/11/2023] Open
Abstract
Triple negative breast cancer (TNBC) is an aggressive disease that is difficult to treat and portends a poor prognosis in many patients. Recent efforts to implement immune checkpoint inhibitors into the treatment landscape of TNBC have led to improved outcomes in a subset of patients both in the early stage and metastatic settings. However, a large portion of patients with TNBC remain resistant to immune checkpoint inhibitors and have limited treatment options beyond cytotoxic chemotherapy. The interplay between the anti-tumor immune response and tumor metabolism contributes to immunotherapy response in the preclinical setting, and likely in the clinical setting as well. Specifically, tumor glycolysis and lactate production influence the tumor immune microenvironment through creation of metabolic competition with infiltrating immune cells, which impacts response to immune checkpoint blockade. In this review, we will focus on how glucose metabolism within TNBC tumors influences the response to immune checkpoint blockade and potential ways of harnessing this information to improve clinical outcomes.
Collapse
Affiliation(s)
- Ashley Schreier
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY, United States
| | - Roberta Zappasodi
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, United States,Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, United States,Parker Institute for Cancer Immunotherapy, San Francisco, CA, United States
| | - Inna Serganova
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, United States,Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Kristy A. Brown
- Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Sandra Demaria
- Department of Radiation Oncology and Department of Pathology, Weill Cornell Medicine, New York, NY, United States
| | - Eleni Andreopoulou
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY, United States,*Correspondence: Eleni Andreopoulou,
| |
Collapse
|
31
|
Zhang Y, Sun Y, Miao Q, Guo S, Wang Q, Shi T, Guo X, Liu S, Cheng G, Wang C, Zhang R. Serum metabolomics analysis in patients with alcohol dependence. Front Psychiatry 2023; 14:1151200. [PMID: 37139316 PMCID: PMC10150058 DOI: 10.3389/fpsyt.2023.1151200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 03/24/2023] [Indexed: 05/05/2023] Open
Abstract
Objective Alcohol dependence (AD) is a chronic recurrent mental disease caused by long-term drinking. It is one of the most prevalent public health problems. However, AD diagnosis lacks objective biomarkers. This study was aimed to shed some light on potential biomarkers of AD patients by investigating the serum metabolomics profiles of AD patients and the controls. Methods Liquid chromatography-mass spectrometry (LC-MS) was used to detect the serum metabolites of 29 AD patients (AD) and 28 controls. Six samples were set aside as the validation set (Control: n = 3; AD group: n = 3), and the remaining were used as the training set (Control: n = 26; AD group: n = 25). Principal component analysis (PCA) and partial least squares discriminant analysis (PCA-DA) were performed to analyze the training set samples. The metabolic pathways were analyzed using the MetPA database. The signal pathways with pathway impact >0.2, value of p <0.05, and FDR < 0.05 were selected. From the screened pathways, the metabolites whose levels changed by at least 3-fold were screened. The metabolites with no numerical overlap in their concentrations in the AD and the control groups were screened out and verified with the validation set. Results The serum metabolomic profiles of the control and the AD groups were significantly different. We identified six significantly altered metabolic signal pathways, including protein digestion and absorption; alanine, aspartate, and glutamate metabolism; arginine biosynthesis; linoleic acid metabolism; butanoate metabolism; and GABAergic synapse. In these six signal pathways, the levels of 28 metabolites were found to be significantly altered. Of these, the alterations of 11 metabolites changed by at least 3-fold compared to the control group. Of these 11 metabolites, those with no numerical overlap in their concentrations between the AD and the control groups were GABA, 4-hydroxybutanoic acid, L-glutamic acid, citric acid and L-glutamine. Conclusion The metabolite profile of the AD group was significantly different from that of the control group. GABA, 4-hydroxybutanoic acid, L-glutamic acid, citric acid, and L-glutamine could be used as potential diagnostic markers for AD.
Collapse
Affiliation(s)
- Yanjie Zhang
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, China
| | - Yajun Sun
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Scientific Research, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Qin Miao
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Addiction, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Shilong Guo
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Qi Wang
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, China
| | - Tianyuan Shi
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, China
| | - Xinsheng Guo
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, China
| | - Shuai Liu
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, China
| | - Guiding Cheng
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, China
| | - Chuansheng Wang
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, China
- *Correspondence: Chuansheng Wang, ; Ruiling Zhang,
| | - Ruiling Zhang
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, China
- *Correspondence: Chuansheng Wang, ; Ruiling Zhang,
| |
Collapse
|
32
|
AMPK is required for recovery from metabolic stress induced by ultrasound microbubble treatment. iScience 2022; 26:105883. [PMID: 36685038 PMCID: PMC9845798 DOI: 10.1016/j.isci.2022.105883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 09/12/2022] [Accepted: 12/22/2022] [Indexed: 12/30/2022] Open
Abstract
Ultrasound-stimulated microbubble (USMB) treatment is a promising strategy for cancer therapy. USMB promotes drug delivery by sonoporation and enhanced endocytosis, and also impairs cell viability. However, USMB elicits heterogeneous effects on cell viability, with apparently minimal effects on a subset of cells. This suggests that mechanisms of adaptation following USMB allow some cells to survive and/or proliferate. Herein, we used several triple negative breast cancer cells to identify the molecular mechanisms of adaptation to USMB-induced stress. We found that USMB alters steady-state levels of amino acids, glycolytic intermediates, and citric acid cycle intermediates, suggesting that USMB imposes metabolic stress on cells. USMB treatment acutely reduces ATP levels and stimulates the phosphorylation and activation of AMP-activated protein kinase (AMPK). AMPK is required to restore ATP levels and support cell proliferation post-USMB treatment. These results suggest that AMPK and metabolic perturbations are likely determinants of the antineoplastic efficacy of USMB treatment.
Collapse
|
33
|
Penta D, Tripathi P, Rajarajan D, Natesh J, Mondal P, Meeran SM. Diindolylmethane Promotes Metabolic Crisis and Enhances the Efficacy of Centchroman in Breast Cancer: A 1H NMR-Based Approach. ACS OMEGA 2022; 7:43147-43160. [PMID: 36467932 PMCID: PMC9713897 DOI: 10.1021/acsomega.2c05832] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 11/07/2022] [Indexed: 05/20/2023]
Abstract
Diindolylmethane (DIM) is a key metabolite of indole-3-carbinol found in cruciferous vegetables such as broccoli, cauliflower, and cabbage. DIM has been known for its anti-cancerous activity through various mechanisms. Most cancer cells, including triple-negative breast cancer (TNBC), adapt distinct metabolic reprogramming for rapid growth and proliferation. Hence, targeting metabolic dysregulation may provide a favorable therapeutic condition for the treatment of TNBC. Earlier, we found that DIM increases the intracellular accumulation of Centchroman (CC), a potential anticancer agent, thereby enhancing the therapeutic potential of CC against breast cancer. However, the role of DIM in regulating TNBC cellular metabolism remains unknown. In the current study, we investigated the potential therapeutic interventions of DIM in TNBC and its metabolic reprogramming in enhancing the efficacy of CC. We found that DIM induced metabolic catastrophe in TNBC cells by regulating aerobic glycolysis and intermediate metabolism. Further, the DIM and CC combination significantly inhibited the TNBC tumor growth in the 4T1-syngeneic model. The inhibition of tumor growth was associated with the downregulation of key aerobic glycolysis mediators such as PKM2, GLUT1, and hypoxia-inducible factor 1α (HIF-1α). This is a first-of-a-kind investigation linking DIM with aerobic glycolysis regulation and enhancing the treatment efficacy of CC against TNBC. Therefore, these findings suggest that DIM-based nutraceuticals and functional foods can be developed as adjuvant therapy for treating metabolically dysregulated TNBC.
Collapse
Affiliation(s)
- Dhanamjai Penta
- Department
of Biochemistry, CSIR-Central Food Technological
Research Institute, Mysore, Karnataka 570020, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Pratima Tripathi
- Plantation
Products, Spices & Flavour Technology, CSIR-Central Food Technological Research Institute, Mysore, Karnataka 570020, India
| | - Dheeran Rajarajan
- Department
of Biochemistry, CSIR-Central Food Technological
Research Institute, Mysore, Karnataka 570020, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Jagadish Natesh
- Department
of Biochemistry, CSIR-Central Food Technological
Research Institute, Mysore, Karnataka 570020, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Priya Mondal
- Department
of Biochemistry, CSIR-Central Food Technological
Research Institute, Mysore, Karnataka 570020, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Syed Musthapa Meeran
- Department
of Biochemistry, CSIR-Central Food Technological
Research Institute, Mysore, Karnataka 570020, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
- , . Phone: +91 821 2517760
ext.: 2476. Fax: +91 821 2516308
| |
Collapse
|
34
|
Kim DH, Kim JS, Mok CS, Chang EH, Choi J, Lim J, Kim CH, Park AR, Bae YJ, Koo BS, Lee HC. dTMP imbalance through thymidylate 5'-phosphohydrolase activity induces apoptosis in triple-negative breast cancers. Sci Rep 2022; 12:20027. [PMID: 36414668 PMCID: PMC9681768 DOI: 10.1038/s41598-022-24706-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022] Open
Abstract
Immunotherapy has a number of advantages over traditional anti-tumor therapy but can cause severe adverse reactions due to an overactive immune system. In contrast, a novel metabolic treatment approach can induce metabolic vulnerability through multiple cancer cell targets. Here, we show a therapeutic effect by inducing nucleotide imbalance and apoptosis in triple negative breast cancer cells (TNBC), by treating with cytosolic thymidylate 5'-phosphohydrolase (CT). We show that a sustained consumption of dTMP by CT could induce dNTP imbalance, leading to apoptosis as tricarboxylic acid cycle intermediates were depleted to mitigate this imbalance. These cytotoxic effects appeared to be different, depending on substrate specificity of the 5' nucleotide or metabolic dependency of the cancer cell lines. Using representative TNBC cell lines, we reveal how the TNBC cells were affected by CT-transfection through extracellular acidification rate (ECAR)/oxygen consumption rate (OCR) analysis and differential transcription/expression levels. We suggest a novel approach for treating refractory TNBC by an mRNA drug that can exploit metabolic dependencies to exacerbate cell metabolic vulnerability.
Collapse
Affiliation(s)
- Dae-Ho Kim
- Research Center, BPgene Co, Ltd, Seoul, 03127 Republic of Korea ,grid.251916.80000 0004 0532 3933Department of Molecular Science and Technology, Ajou University, Suwon, 16499 Republic of Korea ,grid.251916.80000 0004 0532 3933Department of Otolaryngology, Ajou University School of Medicine, Suwon, 16499 Republic of Korea
| | - Jin-Sook Kim
- Research Center, BPgene Co, Ltd, Seoul, 03127 Republic of Korea
| | - Chang-Soo Mok
- Research Center, BPgene Co, Ltd, Seoul, 03127 Republic of Korea ,grid.255168.d0000 0001 0671 5021Department of Life Science, Dongguk University Biomedi Campus, Gyeonggi-do, 10326 Republic of Korea
| | - En-Hyung Chang
- Research Center, BPgene Co, Ltd, Seoul, 03127 Republic of Korea
| | - Jiwon Choi
- Research Center, BPgene Co, Ltd, Seoul, 03127 Republic of Korea
| | - Junsub Lim
- Research Center, BPgene Co, Ltd, Seoul, 03127 Republic of Korea
| | - Chul-Ho Kim
- grid.251916.80000 0004 0532 3933Department of Otolaryngology, Ajou University School of Medicine, Suwon, 16499 Republic of Korea
| | | | | | - Bong-Seong Koo
- Research Center, BPgene Co, Ltd, Seoul, 03127 Republic of Korea
| | - Hyeon-Cheol Lee
- Research Center, BPgene Co, Ltd, Seoul, 03127 Republic of Korea
| |
Collapse
|
35
|
Hu Y, Wu C, Chen Q, Zhang Y, Chen Z. Hydrogen Peroxide Enhances Fatty Acid 2-Hydroxylase Expression to Impede the Lipopolysaccharides-Triggered Apoptosis of Human Mesenchymal Stem Cells. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.3155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
As a type of stem cells that mainly exist in the connective tissue or interstitium, mesenchymal stem cells (MSCs) exhibit great potential in self-renewal and multi-directional differentiation. They have been clinically utilized for the treatment of various diseases including cancer.
This study aims to provide solid evidence for the further development and application of MSCs in human diseases. MSCs were assigned into 5 groups: control group, LPS group, low-, medium- and high-dose hydrogen peroxide groups. After one-hour treatment with LPS, MSCs were exposed to H2O2
for 12 hours followed by analysis of cell apoptosis, viability via EdU staining, TUNEL assay and flow cytometry, FA2H expression by qPCR and Western blotting. The hydrogen peroxide treatment reduced proportion of apoptotic cells induced by LPS, along with enhanced viability and milder DNA
damage. In addition, hydrogen peroxide impeded the LPS-triggered apoptosis of human MSCs. The results above proved that hydrogen peroxide significantly impeded the LPS-triggered apoptosis of MSCs, and further increased cell viability. This protective effect of hydrogen peroxide was mainly
achieved by upregulation of FA2H expression. In conclusion, hydrogen peroxide can enhance FA2H expression to impede the LPS-triggered apoptosis of human MSCs. This finding helps to improve the further development and application of MSCs in treating human diseases.
Collapse
Affiliation(s)
- Yunli Hu
- Department of Cardiovascular Division, Chongqing Jiangjin District Central Hospital, Chongqing, 402260, China
| | - Chunfeng Wu
- Department of Cardiovascular Division, Chongqing Jiangjin District Central Hospital, Chongqing, 402260, China
| | - Qingmei Chen
- Department of Cardiovascular Division, Chongqing Jiangjin District Central Hospital, Chongqing, 402260, China
| | - Yu Zhang
- Department of Cardiovascular Division, Chongqing Jiangjin District Central Hospital, Chongqing, 402260, China
| | - Zhongxia Chen
- Department of Cardiovascular Division, Chongqing Jiangjin District Central Hospital, Chongqing, 402260, China
| |
Collapse
|
36
|
Agostini M, Mancini M, Candi E. Long non-coding RNAs affecting cell metabolism in cancer. Biol Direct 2022; 17:26. [PMID: 36182907 PMCID: PMC9526990 DOI: 10.1186/s13062-022-00341-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 11/27/2021] [Indexed: 11/10/2022] Open
Abstract
Metabolic reprogramming is commonly recognized as one important hallmark of cancers. Cancer cells present significant alteration of glucose metabolism, oxidative phosphorylation, and lipid metabolism. Recent findings demonstrated that long non-coding RNAs control cancer development and progression by modulating cell metabolism. Here, we give an overview of breast cancer metabolic reprogramming and the role of long non-coding RNAs in driving cancer-specific metabolic alteration.
Collapse
Affiliation(s)
- Massimiliano Agostini
- Department Experimental Medicine, University of Rome "Tor Vergata", TOR, Via Montpellier,1, 00133, Rome, Italy
| | - Mara Mancini
- IDI-IRCCS, Via Monti di Creta 104, 00166, Rome, Italy
| | - Eleonora Candi
- Department Experimental Medicine, University of Rome "Tor Vergata", TOR, Via Montpellier,1, 00133, Rome, Italy. .,IDI-IRCCS, Via Monti di Creta 104, 00166, Rome, Italy.
| |
Collapse
|
37
|
Poddar A, Rao SR, Prithviraj P, Kannourakis G, Jayachandran A. Crosstalk between Immune Checkpoint Modulators, Metabolic Reprogramming and Cellular Plasticity in Triple-Negative Breast Cancer. Curr Oncol 2022; 29:6847-6863. [PMID: 36290817 PMCID: PMC9601266 DOI: 10.3390/curroncol29100540] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/16/2022] [Accepted: 09/21/2022] [Indexed: 01/13/2023] Open
Abstract
Breast cancer is one of the major causes of mortality in women worldwide. Accounting for 15-20% of all breast cancer diagnoses, the triple-negative breast cancer (TNBC) subtype presents with an aggressive clinical course, heightened metastatic potential and the poorest short-term prognosis. TNBC does not respond to hormonal therapy, only partially responds to radio- and chemotherapy, and has limited targeted therapy options, thus underlining the critical need for better therapeutic treatments. Although immunotherapy based on immune checkpoint inhibition is emerging as a promising treatment option for TNBC patients, activation of cellular plasticity programs such as metabolic reprogramming (MR) and epithelial-to-mesenchymal transition (EMT) causes immunotherapy to fail. In this report, we review the role of MR and EMT in immune checkpoint dysregulation in TNBCs and specifically shed light on development of novel combination treatment modalities for this challenging disease. We highlight the clinical relevance of crosstalk between MR, EMT, and immune checkpoints in TNBCs.
Collapse
Affiliation(s)
- Arpita Poddar
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3350, Australia
- Department of Materials Science and Engineering, Monash University, Melbourne, VIC 3800, Australia
- Ian Potter NanoBiosensing Facility, NanoBiotechnology Research Laboratory (NBRL), School of Science, RMIT University, Melbourne, VIC 3000, Australia
| | - Sushma R. Rao
- Proteomics, Metabolomics and MS-Imaging Facility, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Prashanth Prithviraj
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3350, Australia
- School of Science, Psychology and Sports, Federation University Australia, Ballarat, VIC 3350, Australia
| | - George Kannourakis
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3350, Australia
- School of Science, Psychology and Sports, Federation University Australia, Ballarat, VIC 3350, Australia
| | - Aparna Jayachandran
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3350, Australia
- School of Science, Psychology and Sports, Federation University Australia, Ballarat, VIC 3350, Australia
- Gallipoli Medical Research Institute, Greenslopes Private Hospital, Brisbane, QLD 4120, Australia
| |
Collapse
|
38
|
Amaro F, Pisoeiro C, Valente MJ, Bastos MDL, Guedes de Pinho P, Carvalho M, Pinto J. Sunitinib versus Pazopanib Dilemma in Renal Cell Carcinoma: New Insights into the In Vitro Metabolic Impact, Efficacy, and Safety. Int J Mol Sci 2022; 23:9898. [PMID: 36077297 PMCID: PMC9456255 DOI: 10.3390/ijms23179898] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/26/2022] [Accepted: 08/26/2022] [Indexed: 11/30/2022] Open
Abstract
Sunitinib and pazopanib are tyrosine kinase inhibitors (TKIs) used as first-line therapy for metastatic renal cell carcinoma (RCC). Although these TKIs are associated with similar survival outcomes, some differences have been reported in their safety profiles. In this work, traditional toxicological endpoints (cell viability and growth, oxidative stress, and nuclear morphology) and 1H NMR spectroscopy-based metabolomics analysis were used to provide new insights into the cytotoxicity and metabolic mechanisms underlying sunitinib and pazopanib treatments. Tumoral (Caki-1) and non-tumoral (HK-2) human renal cells were exposed to clinically relevant concentrations of sunitinib (2 µM) or pazopanib (50 µM). Sunitinib showed selectivity for cancer cells, inhibiting proliferation, and inducing apoptotic death of Caki-1 cells, whereas pazopanib had a similar cytotoxic effect in both tumoral and non-tumoral cells. 1H-NMR metabolomics unveiled a higher impact of sunitinib on the levels of intracellular metabolites of Caki-1 cells (seven dysregulated metabolites), suggesting dysregulations on amino acid, glutathione and glycerophospholipid metabolisms. In contrast, pazopanib had a higher impact on the levels of extracellular metabolites of Caki-1 cells (seven dysregulated metabolites in culture medium), unveiling alterations on amino acid and energetic metabolisms. In HK-2 cells, sunitinib caused only a minor increase in intracellular isoleucine levels, whereas pazopanib induced several alterations on the intracellular (three dysregulated metabolites) and extracellular (three dysregulated metabolites) compartments suggesting changes on amino acid, glycerophospholipid, and energy metabolisms. Our results demonstrate that these TKIs elicit distinct cellular and metabolic responses, with sunitinib showing better in vitro efficacy against target RCC cells and lesser nephrotoxic potential than pazopanib.
Collapse
Affiliation(s)
- Filipa Amaro
- Associate Laboratory i4HB, Department of Biological Sciences, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- UCIBIO-REQUIMTE, Department of Biological Sciences, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Carolina Pisoeiro
- Associate Laboratory i4HB, Department of Biological Sciences, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- UCIBIO-REQUIMTE, Department of Biological Sciences, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Maria João Valente
- National Food Institute, Technical University of Denmark, Kongens Lyngby, 2800 Copenhagen, Denmark
| | - Maria de Lourdes Bastos
- Associate Laboratory i4HB, Department of Biological Sciences, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- UCIBIO-REQUIMTE, Department of Biological Sciences, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Paula Guedes de Pinho
- Associate Laboratory i4HB, Department of Biological Sciences, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- UCIBIO-REQUIMTE, Department of Biological Sciences, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Márcia Carvalho
- Associate Laboratory i4HB, Department of Biological Sciences, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- UCIBIO-REQUIMTE, Department of Biological Sciences, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- FP-I3ID, FP-BHS, University Fernando Pessoa, 4200-150 Porto, Portugal
- Faculty of Health Sciences, University Fernando Pessoa, 4200-150 Porto, Portugal
| | - Joana Pinto
- Associate Laboratory i4HB, Department of Biological Sciences, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- UCIBIO-REQUIMTE, Department of Biological Sciences, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
39
|
Quek LE, van Geldermalsen M, Guan YF, Wahi K, Mayoh C, Balaban S, Pang A, Wang Q, Cowley MJ, Brown KK, Turner N, Hoy AJ, Holst J. Glutamine addiction promotes glucose oxidation in triple-negative breast cancer. Oncogene 2022; 41:4066-4078. [PMID: 35851845 PMCID: PMC9391225 DOI: 10.1038/s41388-022-02408-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 06/30/2022] [Accepted: 07/01/2022] [Indexed: 11/30/2022]
Abstract
Glutamine is a conditionally essential nutrient for many cancer cells, but it remains unclear how consuming glutamine in excess of growth requirements confers greater fitness to glutamine-addicted cancers. By contrasting two breast cancer subtypes with distinct glutamine dependencies, we show that glutamine-indispensable triple-negative breast cancer (TNBC) cells rely on a non-canonical glutamine-to-glutamate overflow, with glutamine carbon routed once through the TCA cycle. Importantly, this single-pass glutaminolysis increases TCA cycle fluxes and replenishes TCA cycle intermediates in TNBC cells, a process that achieves net oxidation of glucose but not glutamine. The coupling of glucose and glutamine catabolism appears hard-wired via a distinct TNBC gene expression profile biased to strip and then sequester glutamine nitrogen, but hampers the ability of TNBC cells to oxidise glucose when glutamine is limiting. Our results provide a new understanding of how metabolically rigid TNBC cells are sensitive to glutamine deprivation and a way to select vulnerable TNBC subtypes that may be responsive to metabolic-targeted therapies.
Collapse
Affiliation(s)
- Lake-Ee Quek
- School of Mathematics and Statistics, The University of Sydney, Camperdown, NSW, Australia.
| | - Michelle van Geldermalsen
- Origins of Cancer Program, Centenary Institute, The University of Sydney, Camperdown, NSW, Australia
- Sydney Medical School, The University of Sydney, Camperdown, NSW, Australia
| | - Yi Fang Guan
- School of Medical Sciences and School of Clinical Medicine, UNSW Sydney, Kensington, NSW, Australia
| | - Kanu Wahi
- School of Medical Sciences and School of Clinical Medicine, UNSW Sydney, Kensington, NSW, Australia
| | - Chelsea Mayoh
- Children's Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington, NSW, Australia
- School of Women's and Children's Health, UNSW Sydney, Kensington, NSW, Australia
| | - Seher Balaban
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia
| | - Angel Pang
- School of Medical Sciences and School of Clinical Medicine, UNSW Sydney, Kensington, NSW, Australia
| | - Qian Wang
- School of Medical Sciences and School of Clinical Medicine, UNSW Sydney, Kensington, NSW, Australia
| | - Mark J Cowley
- Children's Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington, NSW, Australia
- School of Women's and Children's Health, UNSW Sydney, Kensington, NSW, Australia
| | - Kristin K Brown
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Melbourne, VIC, Australia
| | - Nigel Turner
- Department of Pharmacology, School of Medical Sciences, UNSW Sydney, Kensington, NSW, Australia
| | - Andrew J Hoy
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia
| | - Jeff Holst
- School of Medical Sciences and School of Clinical Medicine, UNSW Sydney, Kensington, NSW, Australia.
| |
Collapse
|
40
|
Olelewe C, Kim JH, Ofori S, Mertens RT, Gukathasan S, Awuah SG. Gold(III)-P-chirogenic complex induces mitochondrial dysfunction in triple-negative breast cancer. iScience 2022; 25:104340. [PMID: 35602949 PMCID: PMC9117869 DOI: 10.1016/j.isci.2022.104340] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 03/14/2022] [Accepted: 04/27/2022] [Indexed: 12/19/2022] Open
Abstract
Chemical agents that specifically exploit metabolic vulnerabilities of cancer cells will be beneficial but are rare. The role of oxidative phosphorylation (OXPHOS) in promoting and maintaining triple-negative breast cancer (TNBC) growth provides new treatment opportunity. In this work, we describe AuPhos-19, a small-molecule gold(III)-based agent bearing a chiral phosphine ligand that selectively disrupts mitochondrial metabolism in murine and human TNBC cells but not normal epithelial cells. AuPhos-19 induces potent cytotoxic effect with half maximal inhibitory concentration (IC50) in the nanomolar range (220-650 nM) across different TNBC cell lines. The lipophilic cationic character of AuPhos-19 facilitates interaction with mitochondrial OXPHOS. AuPhos-19 inhibits mitochondria respiration and induces significant AMPK activation. Depolarization of the mitochondria membrane, mitochondria ROS accumulation, and mitochondria DNA depletion provided further indication that AuPhos-19 perturbs mitochondria function. AuPhos-19 inhibits tumor growth in tumor-bearing mice. This study highlights the development of gold-based compounds targeting mitochondrial pathways for efficacious cancer treatment.
Collapse
Affiliation(s)
- Chibuzor Olelewe
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA
| | - Jong Hyun Kim
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA
| | - Samuel Ofori
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA
| | - Randall T. Mertens
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA
| | | | - Samuel G. Awuah
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA
- Center for Pharmaceutical Research and Innovation, College of Pharmacy and Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
41
|
Kokila N, Mahesh B, Roopa K, Daruka Prasad B, Raj K, Manjula S, Mruthunjaya K, Ramu R. Thunbergia mysorensis mediated nano silver oxide for enhanced antibacterial, antioxidant, anticancer potential and in vitro hemolysis evaluation. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.132455] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
42
|
Costa B, Vale N. Drug Metabolism for the Identification of Clinical Biomarkers in Breast Cancer. Int J Mol Sci 2022; 23:3181. [PMID: 35328602 PMCID: PMC8951384 DOI: 10.3390/ijms23063181] [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: 02/17/2022] [Revised: 03/09/2022] [Accepted: 03/15/2022] [Indexed: 02/04/2023] Open
Abstract
Breast cancer is classified into four major molecular subtypes, and is considered a heterogenous disease. The risk profiles and treatment of breast cancer differ according to these subtypes. Early detection dramatically improves the prospects of successful treatment, resulting in a reduction in overall mortality rates. However, almost 30% of women primarily diagnosed with the early-stage disease will eventually develop metastasis or resistance to chemotherapies. Immunotherapies are among the most promising cancer treatment options; however, long-term clinical benefit has only been observed in a small subset of responding patients. The current strategies for diagnosis and treatment rely heavily on histopathological examination and molecular diagnosis, disregarding the tumor microenvironment and microbiome involving cancer cells. In this review, we aim to praise the use of pharmacogenomics and pharmacomicrobiomics as a strategy to identify potential biomarkers for guiding and monitoring therapy in real-time. The finding of these biomarkers can be performed by studying the metabolism of drugs, more specifically, immunometabolism, and its relationship with the microbiome, without neglecting the information provided by genetics. A larger understanding of cancer biology has the potential to improve patient care, enable clinical decisions, and deliver personalized medicine.
Collapse
Affiliation(s)
- Bárbara Costa
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal;
| | - Nuno Vale
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal;
- Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
- Associate Laboratory RISE-Health Research Network, Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| |
Collapse
|
43
|
Zamberlan M, Boeckx A, Muller F, Vinelli F, Ek O, Vianello C, Coart E, Shibata K, Christian A, Grespi F, Giacomello M, Struman I, Scorrano L, Herkenne S. Inhibition of the mitochondrial protein Opa1 curtails breast cancer growth. J Exp Clin Cancer Res 2022; 41:95. [PMID: 35279198 PMCID: PMC8917763 DOI: 10.1186/s13046-022-02304-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 02/26/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Mitochondrial fusion and fission proteins have been nominated as druggable targets in cancer. Whether their inhibition is efficacious in triple negative breast cancer (TNBC) that almost invariably develops chemoresistance is unknown. METHODS We used a combination of bioinformatics analyses of cancer genomic databases, genetic and pharmacological Optic Atrophy 1 (OPA1) inhibition, mitochondrial function and morphology measurements, micro-RNA (miRNA) profiling and formal epistatic analyses to address the role of OPA1 in TNBC proliferation, migration, and invasion in vitro and in vivo. RESULTS We identified a signature of OPA1 upregulation in breast cancer that correlates with worse prognosis. Accordingly, OPA1 inhibition could reduce breast cancer cells proliferation, migration, and invasion in vitro and in vivo. Mechanistically, while OPA1 silencing did not reduce mitochondrial respiration, it increased levels of miRNAs of the 148/152 family known to inhibit tumor growth and invasiveness. Indeed, these miRNAs were epistatic to OPA1 in the regulation of TNBC cells growth and invasiveness. CONCLUSIONS Our data show that targeted inhibition of the mitochondrial fusion protein OPA1 curtails TNBC growth and nominate OPA1 as a druggable target in TNBC.
Collapse
Affiliation(s)
- Margherita Zamberlan
- Department of Biology, University of Padova, Via U. Bassi 58B, 35121, Padova, Italy
- Veneto Institute of Molecular Medicine, Via Orus 2, 35129, Padova, Italy
| | - Amandine Boeckx
- Laboratory of molecular angiogenesis, GIGA-Research, Avenue de l'Hôpital, 1, 4020, Liège, Belgium
| | - Florian Muller
- Laboratory of molecular angiogenesis, GIGA-Research, Avenue de l'Hôpital, 1, 4020, Liège, Belgium
| | - Federica Vinelli
- Department of Biology, University of Padova, Via U. Bassi 58B, 35121, Padova, Italy
- Veneto Institute of Molecular Medicine, Via Orus 2, 35129, Padova, Italy
| | - Olivier Ek
- Department of Biology, University of Padova, Via U. Bassi 58B, 35121, Padova, Italy
| | - Caterina Vianello
- Department of Biology, University of Padova, Via U. Bassi 58B, 35121, Padova, Italy
| | - Emeline Coart
- Laboratory of molecular angiogenesis, GIGA-Research, Avenue de l'Hôpital, 1, 4020, Liège, Belgium
| | - Keitaro Shibata
- Department of Biology, University of Padova, Via U. Bassi 58B, 35121, Padova, Italy
- Veneto Institute of Molecular Medicine, Via Orus 2, 35129, Padova, Italy
| | - Aurélie Christian
- Laboratory of molecular angiogenesis, GIGA-Research, Avenue de l'Hôpital, 1, 4020, Liège, Belgium
| | - Francesca Grespi
- Department of Biology, University of Padova, Via U. Bassi 58B, 35121, Padova, Italy
- Veneto Institute of Molecular Medicine, Via Orus 2, 35129, Padova, Italy
| | - Marta Giacomello
- Department of Biology, University of Padova, Via U. Bassi 58B, 35121, Padova, Italy
| | - Ingrid Struman
- Laboratory of molecular angiogenesis, GIGA-Research, Avenue de l'Hôpital, 1, 4020, Liège, Belgium
| | - Luca Scorrano
- Department of Biology, University of Padova, Via U. Bassi 58B, 35121, Padova, Italy.
- Veneto Institute of Molecular Medicine, Via Orus 2, 35129, Padova, Italy.
| | - Stéphanie Herkenne
- Laboratory of molecular angiogenesis, GIGA-Research, Avenue de l'Hôpital, 1, 4020, Liège, Belgium.
| |
Collapse
|
44
|
Campioni G, Pasquale V, Busti S, Ducci G, Sacco E, Vanoni M. An Optimized Workflow for the Analysis of Metabolic Fluxes in Cancer Spheroids Using Seahorse Technology. Cells 2022; 11:cells11050866. [PMID: 35269488 PMCID: PMC8909358 DOI: 10.3390/cells11050866] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/25/2022] [Accepted: 03/01/2022] [Indexed: 12/12/2022] Open
Abstract
Three-dimensional cancer models, such as spheroids, are increasingly being used to study cancer metabolism because they can better recapitulate the molecular and physiological aspects of the tumor architecture than conventional monolayer cultures. Although Agilent Seahorse XFe96 (Agilent Technologies, Santa Clara, CA, United States) is a valuable technology for studying metabolic alterations occurring in cancer cells, its application to three-dimensional cultures is still poorly optimized. We present a reliable and reproducible workflow for the Seahorse metabolic analysis of three-dimensional cultures. An optimized protocol enables the formation of spheroids highly regular in shape and homogenous in size, reducing variability in metabolic parameters among the experimental replicates, both under basal and drug treatment conditions. High-resolution imaging allows the calculation of the number of viable cells in each spheroid, the normalization of metabolic parameters on a per-cell basis, and grouping of the spheroids as a function of their size. Multivariate statistical tests on metabolic parameters determined by the Mito Stress test on two breast cancer cell lines show that metabolic differences among the studied spheroids are mostly related to the cell line rather than to the size of the spheroid. The optimized workflow allows high-resolution metabolic characterization of three-dimensional cultures, their comparison with monolayer cultures, and may aid in the design and interpretation of (multi)drug protocols.
Collapse
Affiliation(s)
- Gloria Campioni
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy; (G.C.); (V.P.); (S.B.); (G.D.); (E.S.)
- SYSBIO (Centre of Systems Biology), ISBE (Infrastructure Systems Biology Europe), 20126 Milan, Italy
| | - Valentina Pasquale
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy; (G.C.); (V.P.); (S.B.); (G.D.); (E.S.)
- SYSBIO (Centre of Systems Biology), ISBE (Infrastructure Systems Biology Europe), 20126 Milan, Italy
| | - Stefano Busti
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy; (G.C.); (V.P.); (S.B.); (G.D.); (E.S.)
- SYSBIO (Centre of Systems Biology), ISBE (Infrastructure Systems Biology Europe), 20126 Milan, Italy
| | - Giacomo Ducci
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy; (G.C.); (V.P.); (S.B.); (G.D.); (E.S.)
- SYSBIO (Centre of Systems Biology), ISBE (Infrastructure Systems Biology Europe), 20126 Milan, Italy
| | - Elena Sacco
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy; (G.C.); (V.P.); (S.B.); (G.D.); (E.S.)
- SYSBIO (Centre of Systems Biology), ISBE (Infrastructure Systems Biology Europe), 20126 Milan, Italy
| | - Marco Vanoni
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy; (G.C.); (V.P.); (S.B.); (G.D.); (E.S.)
- SYSBIO (Centre of Systems Biology), ISBE (Infrastructure Systems Biology Europe), 20126 Milan, Italy
- Correspondence: ; Tel.: +39-02-6448-3525
| |
Collapse
|
45
|
Ren Z, Liang H, Galbo PM, Dharmaratne M, Kulkarni AS, Fard AT, Aoun ML, Martinez-Lopez N, Suyama K, Benard O, Zheng W, Liu Y, Albanese J, Zheng D, Mar JC, Singh R, Prystowsky MB, Norton L, Hazan RB. Redox signaling by glutathione peroxidase 2 links vascular modulation to metabolic plasticity of breast cancer. Proc Natl Acad Sci U S A 2022; 119:e2107266119. [PMID: 35193955 PMCID: PMC8872779 DOI: 10.1073/pnas.2107266119] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 12/23/2021] [Indexed: 02/07/2023] Open
Abstract
In search of redox mechanisms in breast cancer, we uncovered a striking role for glutathione peroxidase 2 (GPx2) in oncogenic signaling and patient survival. GPx2 loss stimulates malignant progression due to reactive oxygen species/hypoxia inducible factor-α (HIF1α)/VEGFA (vascular endothelial growth factor A) signaling, causing poor perfusion and hypoxia, which were reversed by GPx2 reexpression or HIF1α inhibition. Ingenuity Pathway Analysis revealed a link between GPx2 loss, tumor angiogenesis, metabolic modulation, and HIF1α signaling. Single-cell RNA analysis and bioenergetic profiling revealed that GPx2 loss stimulated the Warburg effect in most tumor cell subpopulations, except for one cluster, which was capable of oxidative phosphorylation and glycolysis, as confirmed by coexpression of phosphorylated-AMPK and GLUT1. These findings underscore a unique role for redox signaling by GPx2 dysregulation in breast cancer, underlying tumor heterogeneity, leading to metabolic plasticity and malignant progression.
Collapse
Affiliation(s)
- Zuen Ren
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Huizhi Liang
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Phillip M Galbo
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Malindrie Dharmaratne
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, 4072 QLD, Australia
| | - Ameya S Kulkarni
- Department of Endocrinology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Atefeh Taherian Fard
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, 4072 QLD, Australia
| | - Marie Louise Aoun
- Department of Endocrinology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Nuria Martinez-Lopez
- Department of Endocrinology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Kimita Suyama
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461
| | | | - Wei Zheng
- Department of Hematology and Medical Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Yang Liu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461
- Department of Neurology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Joseph Albanese
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461
- Department of Neurology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Jessica C Mar
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, 4072 QLD, Australia
| | - Rajat Singh
- Department of Endocrinology, Albert Einstein College of Medicine, Bronx, NY 10461
| | | | - Larry Norton
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY 10021
| | - Rachel B Hazan
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461;
| |
Collapse
|
46
|
Williams SD, Sakwe AM. Reduced Expression of Annexin A6 Induces Metabolic Reprogramming That Favors Rapid Fatty Acid Oxidation in Triple-Negative Breast Cancer Cells. Cancers (Basel) 2022; 14:1108. [PMID: 35267416 PMCID: PMC8909273 DOI: 10.3390/cancers14051108] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/17/2022] [Accepted: 02/20/2022] [Indexed: 12/01/2022] Open
Abstract
The ability of cancer cells to alter their metabolism is one of the major mechanisms underlying rapid tumor progression and/or therapeutic resistance in solid tumors, including the hard-to-treat triple-negative breast cancer (TNBC) subtype. Here, we assessed the contribution of the tumor suppressor, Annexin A6 (AnxA6), in the metabolic adaptation of basal-like (AnxA6-low) versus mesenchymal-like (AnxA6-high), as well as in lapatinib-resistant TNBC cells. Using model basal-like and mesenchymal-like TNBC cell lines, we show that TNBC cells also exhibit metabolic heterogeneity. The downregulation of AnxA6 in TNBC cells generally attenuated mitochondrial respiration, glycolytic flux, and cellular ATP production capacity resulting in a quiescent metabolic phenotype. We also show that AnxA6 depletion in mesenchymal-like TNBC cells was associated with a rapid uptake and mitochondrial fatty acid oxidation and diminished lipid droplet accumulation and altered the lipogenic metabolic phenotype of these cells to a lypolytic metabolic phenotype. The overexpression or chronic lapatinib-induced upregulation of AnxA6 in AnxA6-low TNBC cells reversed the quiescent/lypolytic phenotype to a more lipogenic/glycolytic phenotype with gluconeogenic precursors as additional metabolites. Collectively, these data suggest that the expression status of AnxA6 in TNBC cells underlies distinct metabolic adaptations of basal-like and mesenchymal-like TNBC subsets in response to cellular stress and/or therapeutic intervention and suggest AnxA6 as a biomarker for metabolic subtyping of TNBC subsets.
Collapse
Affiliation(s)
| | - Amos M. Sakwe
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN 37208, USA;
| |
Collapse
|
47
|
Liao C, Glodowski CR, Fan C, Liu J, Mott KR, Kaushik A, Vu H, Locasale JW, McBrayer SK, DeBerardinis RJ, Perou CM, Zhang Q. Integrated Metabolic Profiling and Transcriptional Analysis Reveals Therapeutic Modalities for Targeting Rapidly Proliferating Breast Cancers. Cancer Res 2022; 82:665-680. [PMID: 34911787 PMCID: PMC8857046 DOI: 10.1158/0008-5472.can-21-2745] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/31/2021] [Accepted: 12/13/2021] [Indexed: 11/16/2022]
Abstract
Metabolic dysregulation is a prominent feature in breast cancer, but it remains poorly characterized in patient tumors. In this study, untargeted metabolomics analysis of triple-negative breast cancer (TNBC) and patient with estrogen receptor (ER)-positive breast cancer samples, as well as TNBC patient-derived xenografts (PDX), revealed two major metabolic groups independent of breast cancer histologic subtypes: a "Nucleotide/Carbohydrate-Enriched" group and a "Lipid/Fatty Acid-Enriched" group. Cell lines grown in vivo more faithfully recapitulated the metabolic profiles of patient tumors compared with those grown in vitro. Integrated metabolic and gene expression analyses identified genes that strongly correlate with metabolic dysregulation and predict patient prognosis. As a proof of principle, targeting Nucleotide/Carbohydrate-Enriched TNBC cell lines or PDX xenografts with a pyrimidine biosynthesis inhibitor or a glutaminase inhibitor led to therapeutic efficacy. In multiple in vivo models of TNBC, treatment with the pyrimidine biosynthesis inhibitor conferred better therapeutic outcomes than chemotherapeutic agents. This study provides a metabolic stratification of breast tumor samples that can guide the selection of effective therapeutic strategies targeting breast cancer subsets. In addition, we have developed a public, interactive data visualization portal (http://brcametab.org) based on the data generated from this study to facilitate future research. SIGNIFICANCE A multiomics strategy that integrates metabolic and gene expression profiling in patient tumor samples and animal models identifies effective pharmacologic approaches to target rapidly proliferating breast tumor subtypes.
Collapse
Affiliation(s)
- Chengheng Liao
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- These authors contributed equally
| | - Cherise Ryan Glodowski
- Department of Pathology and Laboratory Medicine, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
- These authors contributed equally
| | - Cheng Fan
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Juan Liu
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Kevin R. Mott
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Akash Kaushik
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Hieu Vu
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Jason W. Locasale
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Samuel K. McBrayer
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Ralph J. DeBerardinis
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Charles M. Perou
- Department of Pathology and Laboratory Medicine, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
- Department of Genetics, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Qing Zhang
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
48
|
Romashev NF, Abramov PA, Bakaev IV, Fomenko IS, Samsonenko DG, Novikov AS, Tong KKH, Ahn D, Dorovatovskii PV, Zubavichus YV, Ryadun AA, Patutina OA, Sokolov MN, Babak MV, Gushchin AL. Heteroleptic Pd(II) and Pt(II) Complexes with Redox-Active Ligands: Synthesis, Structure, and Multimodal Anticancer Mechanism. Inorg Chem 2022; 61:2105-2118. [PMID: 35029379 DOI: 10.1021/acs.inorgchem.1c03314] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
A series of heteroleptic square-planar Pt and Pd complexes with bis(diisopropylphenyl) iminoacenaphtene (dpp-Bian) and Cl, 1,3-dithia-2-thione-4,5-dithiolate (dmit), or 1,3-dithia-2-thione-4,5-diselenolate (dsit) ligands have been prepared and characterized by spectroscopic techniques, elemental analysis, X-ray diffraction analysis, and cyclic voltammetry (CV). The intermolecular noncovalent interactions in the crystal structures were assessed by density functional theory (DFT) calculations. The anticancer activity of Pd complexes in breast cancer cell lines was limited by their solubility. Pd(dpp-Bian) complexes with dmit and dsit ligands as well as an uncoordinated dpp-Bian ligand were devoid of cytotoxicity, while the [Pd(dpp-Bian)Cl2] complex was cytotoxic. On the contrary, all Pt(dpp-Bian) complexes demonstrated anticancer activity in a low micromolar concentration range, which was 8-20 times higher than the activity of cisplatin, and up to 2.5-fold selectivity toward cancer cells over healthy fibroblasts. The presence of a redox-active dpp-Bian ligand in Pt and Pd complexes resulted in the induction of reactive oxygen species (ROS) in cancer cells. In addition, these complexes were able to intercalate into DNA, indicating the dual mechanism of action.
Collapse
Affiliation(s)
- Nikolai F Romashev
- Nikolaev Institute of Inorganic Chemistry SB RAS, 3 Acad. Lavrentiev Ave., Novosibirsk 630090, Russia
| | - Pavel A Abramov
- Nikolaev Institute of Inorganic Chemistry SB RAS, 3 Acad. Lavrentiev Ave., Novosibirsk 630090, Russia
| | - Ivan V Bakaev
- Nikolaev Institute of Inorganic Chemistry SB RAS, 3 Acad. Lavrentiev Ave., Novosibirsk 630090, Russia
- Novosibirsk State University, 1 Pirogov st., Novosibirsk 630090, Russia
| | - Iakov S Fomenko
- Nikolaev Institute of Inorganic Chemistry SB RAS, 3 Acad. Lavrentiev Ave., Novosibirsk 630090, Russia
| | - Denis G Samsonenko
- Nikolaev Institute of Inorganic Chemistry SB RAS, 3 Acad. Lavrentiev Ave., Novosibirsk 630090, Russia
| | - Alexander S Novikov
- Institute of Chemistry, Saint Petersburg State University, Universitetskaya Nab., 7/9, Saint Petersburg 199034, Russia
| | - Kelvin K H Tong
- Drug Discovery Lab, Department of Chemistry, City University of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, People's Republic of China
| | - Dohyun Ahn
- Drug Discovery Lab, Department of Chemistry, City University of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, People's Republic of China
| | - Pavel V Dorovatovskii
- National Research Center "Kurchatov Institute", Kurchatov Square 1, Moscow 123182, Russia
| | - Yan V Zubavichus
- Boreskov Institute of Catalysis, 5 Acad. Lavrentiev Ave., Novosibirsk 630090, Russia
| | - Aleksey A Ryadun
- Nikolaev Institute of Inorganic Chemistry SB RAS, 3 Acad. Lavrentiev Ave., Novosibirsk 630090, Russia
| | - Olga A Patutina
- Institute of Chemical Biology and Fundamental Medicine, 8 Acad. Lavrentiev Ave., Novosibirsk 630090, Russia
| | - Maxim N Sokolov
- Nikolaev Institute of Inorganic Chemistry SB RAS, 3 Acad. Lavrentiev Ave., Novosibirsk 630090, Russia
| | - Maria V Babak
- Drug Discovery Lab, Department of Chemistry, City University of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, People's Republic of China
| | - Artem L Gushchin
- Nikolaev Institute of Inorganic Chemistry SB RAS, 3 Acad. Lavrentiev Ave., Novosibirsk 630090, Russia
| |
Collapse
|
49
|
Xue D, Xu Y, Kyani A, Roy J, Dai L, Sun D, Neamati N. Discovery and Lead Optimization of Benzene-1,4-disulfonamides as Oxidative Phosphorylation Inhibitors. J Med Chem 2022; 65:343-368. [PMID: 34982568 DOI: 10.1021/acs.jmedchem.1c01509] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Inhibition of oxidative phosphorylation (OXPHOS) is a promising therapeutic strategy for select cancers that are dependent on aerobic metabolism. Here, we report the discovery, optimization, and structure-activity relationship (SAR) study of a series of novel OXPHOS inhibitors. The hit compound, benzene-1,4-disulfonamide 1, was discovered in a phenotypic screen selective for cytotoxicity in a galactose-containing medium. Our multi-parameter optimization campaign led to the discovery of 65 (DX3-235), showing nanomolar inhibition of complex I function and adenosine triphosphate (ATP) production in a galactose-containing medium resulting in significant cytotoxicity. Importantly, 64 (DX3-234), a close analogue of 65, is well tolerated in mice and shows significant single agent efficacy in a Pan02 syngeneic pancreatic cancer model, suggesting that highly potent and selective OXPHOS inhibitors can be useful for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Ding Xue
- Departments of Medicinal Chemistry, University of Michigan, North Campus Research Complex, 1600 Huron Parkway, Ann Arbor, Michigan 48109, United States
| | - Yibin Xu
- Departments of Medicinal Chemistry, University of Michigan, North Campus Research Complex, 1600 Huron Parkway, Ann Arbor, Michigan 48109, United States
| | - Armita Kyani
- Departments of Medicinal Chemistry, University of Michigan, North Campus Research Complex, 1600 Huron Parkway, Ann Arbor, Michigan 48109, United States
| | - Joyeeta Roy
- Departments of Medicinal Chemistry, University of Michigan, North Campus Research Complex, 1600 Huron Parkway, Ann Arbor, Michigan 48109, United States
| | - Lipeng Dai
- Departments of Medicinal Chemistry, University of Michigan, North Campus Research Complex, 1600 Huron Parkway, Ann Arbor, Michigan 48109, United States.,Pharmaceutical Sciences, College of Pharmacy, Rogel Cancer Center, University of Michigan, North Campus Research Complex, 1600 Huron Parkway, Ann Arbor, Michigan 48109, United States
| | - Duxin Sun
- Departments of Medicinal Chemistry, University of Michigan, North Campus Research Complex, 1600 Huron Parkway, Ann Arbor, Michigan 48109, United States.,Pharmaceutical Sciences, College of Pharmacy, Rogel Cancer Center, University of Michigan, North Campus Research Complex, 1600 Huron Parkway, Ann Arbor, Michigan 48109, United States
| | - Nouri Neamati
- Departments of Medicinal Chemistry, University of Michigan, North Campus Research Complex, 1600 Huron Parkway, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
50
|
Yao C, Weng J, Feng L, Zhang W, Xu Y, Zhang P, Tanaka Y, Su L. SIPA1 Enhances Aerobic Glycolysis Through HIF-2α Pathway to Promote Breast Cancer Metastasis. Front Cell Dev Biol 2022; 9:779169. [PMID: 35096814 PMCID: PMC8790513 DOI: 10.3389/fcell.2021.779169] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 12/17/2021] [Indexed: 12/21/2022] Open
Abstract
Increased dependence on aerobic glycolysis is characteristic of most cancer cells, whereas the mechanism underlying the promotion of aerobic glycolysis in metastatic breast cancer cells under ambient oxygen has not been well understood. Here, we demonstrated that aberrant expression of signal-induced proliferation-associated 1 (SIPA1) enhanced aerobic glycolysis and altered the main source of ATP production from oxidative phosphorylation to glycolysis in breast cancer cells. We revealed that SIPA1 promoted the transcription of EPAS1, which is known as the gene encoding hypoxia-inducible factor-2α (HIF-2α) and up-regulated the expression of multiple glycolysis-related genes to increase aerobic glycolysis. We also found that blocking aerobic glycolysis by either knocking down SIPA1 expression or oxamate treatment led to the suppression of tumor metastasis of breast cancer cells both in vitro and in vivo. Taken together, aberrant expression of SIPA1 resulted in the alteration of glucose metabolism from oxidative phosphorylation to aerobic glycolysis even at ambient oxygen levels, which might aggravate the malignancy of breast cancer cells. The present findings indicate a potential target for the development of therapeutics against breast cancers with dysregulated SIPA1 expression.
Collapse
Affiliation(s)
- Chenguang Yao
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Weng
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Lingyun Feng
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Wanjun Zhang
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Xu
- Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Peijing Zhang
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Yoshimasa Tanaka
- Center for Medical Innovation, Nagasaki University, Nagasaki, Japan
| | - Li Su
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|