1
|
Sun Y, Yang X, Kong F, Dong FY, Li N, Wang S. The mechanisms and effects of lactylation modification in different kinds of cancers. Discov Oncol 2025; 16:560. [PMID: 40249419 PMCID: PMC12008107 DOI: 10.1007/s12672-025-02359-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 04/09/2025] [Indexed: 04/19/2025] Open
Abstract
Lactylation, a recently identified post-translational modification, has garnered significant attention for its associations with various diseases, particularly its critical role in tumor progression and treatment. It is emerging as a potential clinical target. The elevated metabolic activity of cancer cells often leads to excessive lactate accumulation, a phenomenon termed the "Warburg effect", which is a hallmark of the tumor microenvironment. Recent research reveals that lactate is not merely a metabolic byproduct but also serves as a substrate for protein lactylation, influencing tumor development by regulating cellular signaling, gene expression, and immune responses. This dual role has become a focal point for scientists and clinicians seeking novel therapeutic strategies targeting lactate-related pathways. Despite growing interest, the detailed mechanisms and therapeutic applications of lactylation across different cancer types remain inadequately explored. This review synthesizes current findings on lactylation mechanisms in various tumors, highlights potential therapeutic targets, and offers new perspectives to advance cancer treatment.
Collapse
Affiliation(s)
- Yixun Sun
- College of Clinical Medicine, Jining Medical University, Jining, 272007, Shandong, China
| | - Xiaxia Yang
- Department of Laboratory Medicine,, Affiliated Hospital of Jining Medical University, Jining Medical University, 89 Guhuai Road, Jining, 272000, Shandong, China
| | - Feifei Kong
- Department of Laboratory Medicine,, Affiliated Hospital of Jining Medical University, Jining Medical University, 89 Guhuai Road, Jining, 272000, Shandong, China
| | - Feng Yun Dong
- Department of Laboratory Medicine,, Affiliated Hospital of Jining Medical University, Jining Medical University, 89 Guhuai Road, Jining, 272000, Shandong, China
| | - Na Li
- Department of Pediatrics, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272007, Shandong, China
| | - Sen Wang
- Department of Laboratory Medicine,, Affiliated Hospital of Jining Medical University, Jining Medical University, 89 Guhuai Road, Jining, 272000, Shandong, China.
| |
Collapse
|
2
|
Bustos G, Ahumada-Castro U, Silva-Pavez E, Huerta H, Puebla A, Quezada C, Morgado-Cáceres P, Casanova-Canelo C, Smith-Cortinez N, Podunavac M, Oyarce C, Lladser A, Farias P, Lovy A, Molgó J, Torres VA, Zakarian A, Cárdenas JC. The IP 3R inhibitor desmethylxestospongin B reduces tumor cell migration, invasion and metastasis by impairing lysosome acidification and β1-integrin recycling. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167557. [PMID: 39486657 DOI: 10.1016/j.bbadis.2024.167557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 09/26/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024]
Abstract
Cancer is the second leading cause of death worldwide. >90 % of cancer-related deaths are due to metastasis, a process that depends on the ability of cancer cells to leave the primary tumor, migrate, and colonize different tissues. Inositol 1,4,5-trisphosphate receptor (IP3R)-mediated Ca2+ signaling plays an essential role in maintaining the homeostasis of cancer cells and the sustained proliferation. Desmethylxestospongin B (dmXeB) is a specific inhibitor of the IP3R that selectively arrests cell proliferation and promotes cancer cell death at high concentrations. However, whether migration, invasion and metastasis can be affected by this drug is unknown. Here, by using the highly metastatic triple negative breast cancer (TNBC) cell line MDA-MB-231, we demonstrate that a prolonged inhibition of IP3R-mediated Ca2+ signals with dmXeB significantly reduces cell migration and invasion in vitro and metastasis in vivo. We found that this phenomenon was independent of the bioenergetic control of IP3R over the mitochondria and AMPK activation. Furthermore, employing a tandem LC3-GFP-mcherry assay, we found that prolonged inhibition of IP3R with dmXeB leads to diminished autophagic flux. This reduction can be attributed to impaired lysosomal acidification, as evidenced by assessments using DQ-BSA and pHrodo. Since cell migration requires appropriate assembly and disassembly of focal adhesions, along with the internalization and recycling of integrins via autophagy, we explored the dependency of integrin recycling from autophagosomes, finding that IP3R inhibition with dmXeB impaired the recycling of β1-integrins, which accumulated within autophagosomes. Our findings reveal an unexpected effect of IP3R inhibition with dmXeB in cancer cells that could represent a novel therapeutic strategy for the treatment of cancer metastasis.
Collapse
Affiliation(s)
- Galdo Bustos
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile; Geroscience Center for Brain Health and Metabolism, Santiago 8580745, Chile
| | - Ulises Ahumada-Castro
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile; Geroscience Center for Brain Health and Metabolism, Santiago 8580745, Chile
| | - Eduardo Silva-Pavez
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile
| | - Hernán Huerta
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile; Geroscience Center for Brain Health and Metabolism, Santiago 8580745, Chile
| | - Andrea Puebla
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile; Geroscience Center for Brain Health and Metabolism, Santiago 8580745, Chile
| | - Camila Quezada
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile; Geroscience Center for Brain Health and Metabolism, Santiago 8580745, Chile
| | - Pablo Morgado-Cáceres
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile; Geroscience Center for Brain Health and Metabolism, Santiago 8580745, Chile
| | - César Casanova-Canelo
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile; Geroscience Center for Brain Health and Metabolism, Santiago 8580745, Chile
| | - Natalia Smith-Cortinez
- Department of Gastroenterology and Hepatology, UMCG, University of Groningen, Netherlands
| | - Maša Podunavac
- Department of Chemistry and Biochemistry, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Cesar Oyarce
- Department of Medical Microbiology and Infection Prevention, Tumor Virology and Cancer Immunotherapy, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Alvaro Lladser
- Centro Cientifico y Tecnologico de Excelencia Ciencia & Vida, Fundación Ciencia and Vida, Chile; Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Paula Farias
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile; Geroscience Center for Brain Health and Metabolism, Santiago 8580745, Chile
| | - Alenka Lovy
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile; Geroscience Center for Brain Health and Metabolism, Santiago 8580745, Chile
| | - Jordi Molgó
- Université Paris-Saclay, CEA, Département Médicaments et Technologies pour la Santé, Service d'Ingénierie Moléculaire pour la Santé (SIMoS), Equipe Mixte de Recherche CNRS 9004, F-91191 Gif-sur-Yvette, France
| | - Vicente A Torres
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Chile; Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santiago 8380453, Chile; Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, Santiago, Chile
| | - Armen Zakarian
- Department of Chemistry and Biochemistry, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - J César Cárdenas
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile; Geroscience Center for Brain Health and Metabolism, Santiago 8580745, Chile; Department of Chemistry and Biochemistry, University of California Santa Barbara, Santa Barbara, CA 93106, USA; The Buck Institute for Research on Aging, Novato, USA.
| |
Collapse
|
3
|
Aye HM, Li FJ, He CY. Dynamic composition of stress granules in Trypanosoma brucei. PLoS Pathog 2024; 20:e1012666. [PMID: 39480887 PMCID: PMC11556693 DOI: 10.1371/journal.ppat.1012666] [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: 03/07/2024] [Revised: 11/12/2024] [Accepted: 10/14/2024] [Indexed: 11/02/2024] Open
Abstract
Stress granules (SGs) are stress-induced RNA condensates consisting of stalled initiation complexes resulting from translational inhibition. The biochemical composition and function of SGs are highly diverse, and this diversity has been attributed to different stress conditions, signalling pathways involved and specific cell types. Interestingly, mRNA decay components, which are found in ubiquitous cytoplasmic foci known as processing bodies (PB), have also been identified in SG proteomes. A major challenge in current SG studies is to understand the cause of SG diversity, as well as the function of SG under different stress conditions. Trypanosoma brucei is a single-cellular parasite that causes Human African Trypanosomiasis (sleeping sickness). In this study, we showed that by varying the supply of extracellular carbon sources during starvation, cellular ATP levels changed rapidly, resulting in SGs of different compositions and dynamics. We identified a subset of SG components, which dissociated from the SGs in response to cellular ATP depletion. Using expansion microscopy, we observed sub-granular compartmentalization of PB- and SG-components within the stress granules. Our results highlight the importance of cellular ATP in SG composition and dynamics, providing functional insight to SGs formed under different stress conditions.
Collapse
Affiliation(s)
- Htay Mon Aye
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Feng-Jun Li
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Cynthia Y. He
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| |
Collapse
|
4
|
Liu Y, Wang YJ, Du Y, Liu W, Huang X, Fan Z, Lu J, Yi R, Xiang XW, Xia X, Gu H, Liu YJ, Liu B. DNA nanomachines reveal an adaptive energy mode in confinement-induced amoeboid migration powered by polarized mitochondrial distribution. Proc Natl Acad Sci U S A 2024; 121:e2317492121. [PMID: 38547056 PMCID: PMC10998588 DOI: 10.1073/pnas.2317492121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 02/19/2024] [Indexed: 04/02/2024] Open
Abstract
Energy metabolism is highly interdependent with adaptive cell migration in vivo. Mechanical confinement is a critical physical cue that induces switchable migration modes of the mesenchymal-to-amoeboid transition (MAT). However, the energy states in distinct migration modes, especially amoeboid-like stable bleb (A2) movement, remain unclear. In this report, we developed multivalent DNA framework-based nanomachines to explore strategical mitochondrial trafficking and differential ATP levels during cell migration in mechanically heterogeneous microenvironments. Through single-particle tracking and metabolomic analysis, we revealed that fast A2-moving cells driven by biomimetic confinement recruited back-end positioning of mitochondria for powering highly polarized cytoskeletal networks, preferentially adopting an energy-saving mode compared with a mesenchymal mode of cell migration. We present a versatile DNA nanotool for cellular energy exploration and highlight that adaptive energy strategies coordinately support switchable migration modes for facilitating efficient metastatic escape, offering a unique perspective for therapeutic interventions in cancer metastasis.
Collapse
Affiliation(s)
- Yixin Liu
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Lab of Molecular Engineering of Polymers, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai200438, China
| | - Ya-Jun Wang
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Lab of Molecular Engineering of Polymers, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai200438, China
| | - Yang Du
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Lab of Molecular Engineering of Polymers, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai200438, China
| | - Wei Liu
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Lab of Molecular Engineering of Polymers, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai200438, China
| | - Xuedong Huang
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Lab of Molecular Engineering of Polymers, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai200438, China
| | - Zihui Fan
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Lab of Molecular Engineering of Polymers, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai200438, China
| | - Jiayin Lu
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Lab of Molecular Engineering of Polymers, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai200438, China
| | - Runqiu Yi
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Lab of Molecular Engineering of Polymers, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai200438, China
| | - Xiao-Wei Xiang
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Lab of Molecular Engineering of Polymers, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai200438, China
| | - Xinwei Xia
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Lab of Molecular Engineering of Polymers, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai200438, China
| | - Hongzhou Gu
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Lab of Molecular Engineering of Polymers, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai200438, China
| | - Yan-Jun Liu
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Lab of Molecular Engineering of Polymers, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai200438, China
| | - Baohong Liu
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Lab of Molecular Engineering of Polymers, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai200438, China
| |
Collapse
|
5
|
O’Callaghan E, Navarrete-Lopez P, Štiavnická M, Sánchez JM, Maroto M, Pericuesta E, Fernández-González R, O’Meara C, Eivers B, Kelleher MM, Evans RD, Mapel XM, Lloret-Villas A, Pausch H, Balastegui-Alarcón M, Avilés M, Sanchez-Rodriguez A, Roldan ERS, McDonald M, Kenny DA, Fair S, Gutiérrez-Adán A, Lonergan P. Adenylate kinase 9 is essential for sperm function and male fertility in mammals. Proc Natl Acad Sci U S A 2023; 120:e2305712120. [PMID: 37812723 PMCID: PMC10589668 DOI: 10.1073/pnas.2305712120] [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: 04/10/2023] [Accepted: 08/23/2023] [Indexed: 10/11/2023] Open
Abstract
Despite passing routine laboratory tests for semen quality, bulls used in artificial insemination exhibit significant variation in fertility. Routine analysis of fertility data identified a dairy bull with extreme subfertility (10% pregnancy rate). To characterize the subfertility phenotype, a range of in vitro, in vivo, and molecular assays were carried out. Sperm from the subfertile bull exhibited reduced motility and severely reduced caffeine-induced hyperactivation compared to controls. Ability to penetrate the zona pellucida, cleavage rate, cleavage kinetics, and blastocyst yield after IVF or AI were significantly lower than in control bulls. Whole-genome sequencing from semen and RNA sequencing of testis tissue revealed a critical mutation in adenylate kinase 9 (AK9) that impaired splicing, leading to a premature termination codon and a severely truncated protein. Mice deficient in AK9 were generated to further investigate the function of the gene; knockout males were phenotypically indistinguishable from their wild-type littermates but produced immotile sperm that were incapable of normal fertilization. These sperm exhibited numerous abnormalities, including a low ATP concentration and reduced motility. RNA-seq analysis of their testis revealed differential gene expression of components of the axoneme and sperm flagellum as well as steroid metabolic processes. Sperm ultrastructural analysis showed a high percentage of sperm with abnormal flagella. Combined bovine and murine data indicate the essential metabolic role of AK9 in sperm motility and/or hyperactivation, which in turn affects sperm binding and penetration of the zona pellucida. Thus, AK9 has been found to be directly implicated in impaired male fertility in mammals.
Collapse
Affiliation(s)
- Elena O’Callaghan
- Animal and Crop Sciences, School of Agriculture and Food Science, University College Dublin, Belfield, DublinD04 V1W8, Ireland
| | - Paula Navarrete-Lopez
- Departamento de Reproducción Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria-Centro Nacional integrado en la Agencia Estatal Consejo Superior de Investigaciones Científicas, Madrid28040, Spain
| | - Miriama Štiavnická
- Department of Biological Sciences, Bernal Institute, Faculty of Science and Engineering, University of Limerick, LimerickV94 T9PX, Ireland
| | - José M. Sánchez
- Animal and Crop Sciences, School of Agriculture and Food Science, University College Dublin, Belfield, DublinD04 V1W8, Ireland
- Departamento de Reproducción Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria-Centro Nacional integrado en la Agencia Estatal Consejo Superior de Investigaciones Científicas, Madrid28040, Spain
| | - Maria Maroto
- Departamento de Reproducción Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria-Centro Nacional integrado en la Agencia Estatal Consejo Superior de Investigaciones Científicas, Madrid28040, Spain
| | - Eva Pericuesta
- Departamento de Reproducción Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria-Centro Nacional integrado en la Agencia Estatal Consejo Superior de Investigaciones Científicas, Madrid28040, Spain
| | - Raul Fernández-González
- Departamento de Reproducción Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria-Centro Nacional integrado en la Agencia Estatal Consejo Superior de Investigaciones Científicas, Madrid28040, Spain
| | - Ciara O’Meara
- National Cattle Breeding Centre, County KildareW91 WF59, Ireland
| | - Bernard Eivers
- National Cattle Breeding Centre, County KildareW91 WF59, Ireland
| | - Margaret M. Kelleher
- Irish Cattle Breeding Federation, Link Road, Ballincollig, County CorkP31 D452, Ireland
| | - Ross D. Evans
- Irish Cattle Breeding Federation, Link Road, Ballincollig, County CorkP31 D452, Ireland
| | - Xena M. Mapel
- Animal Genomics, Institute of Agricultural Sciences, ETH Zürich, Zürich8092, Switzerland
| | - Audald Lloret-Villas
- Animal Genomics, Institute of Agricultural Sciences, ETH Zürich, Zürich8092, Switzerland
| | - Hubert Pausch
- Animal Genomics, Institute of Agricultural Sciences, ETH Zürich, Zürich8092, Switzerland
| | - Miriam Balastegui-Alarcón
- Departamento de Biología Celular e Histología, Universidad de Murcia-Instituto Murciano de Investigación Biosanitaria Pascual Parrilla, Murcia30120, Spain
| | - Manuel Avilés
- Departamento de Biología Celular e Histología, Universidad de Murcia-Instituto Murciano de Investigación Biosanitaria Pascual Parrilla, Murcia30120, Spain
| | - Ana Sanchez-Rodriguez
- Departmento de Biodiversidad y Biología Evolutiva, Museo Nacional de Ciencias Naturales, Madrid28006, Spain
| | - Eduardo R. S. Roldan
- Departmento de Biodiversidad y Biología Evolutiva, Museo Nacional de Ciencias Naturales, Madrid28006, Spain
| | - Michael McDonald
- Animal and Crop Sciences, School of Agriculture and Food Science, University College Dublin, Belfield, DublinD04 V1W8, Ireland
| | - David A. Kenny
- Animal and Bioscience Department, Teagasc, Animal and Grassland Research and Innovation Centre, Grange, Dunsany, County MeathC15 PW93, Ireland
| | - Sean Fair
- Department of Biological Sciences, Bernal Institute, Faculty of Science and Engineering, University of Limerick, LimerickV94 T9PX, Ireland
| | - Alfonso Gutiérrez-Adán
- Departamento de Reproducción Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria-Centro Nacional integrado en la Agencia Estatal Consejo Superior de Investigaciones Científicas, Madrid28040, Spain
| | - Patrick Lonergan
- Animal and Crop Sciences, School of Agriculture and Food Science, University College Dublin, Belfield, DublinD04 V1W8, Ireland
| |
Collapse
|
6
|
Parlani M, Jorgez C, Friedl P. Plasticity of cancer invasion and energy metabolism. Trends Cell Biol 2023; 33:388-402. [PMID: 36328835 PMCID: PMC10368441 DOI: 10.1016/j.tcb.2022.09.009] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 11/05/2022]
Abstract
Energy deprivation is a frequent adverse event in tumors that is caused by mutations, malperfusion, hypoxia, and nutrition deficit. The resulting bioenergetic stress leads to signaling and metabolic adaptation responses in tumor cells, secures survival, and adjusts migration activity. The kinetic responses of cancer cells to energy deficit were recently identified, including a switch of invasive cancer cells to energy-conservative amoeboid migration and an enhanced capability for distant metastasis. We review the energy programs employed by different cancer invasion modes including collective, mesenchymal, and amoeboid migration, as well as their interconversion in response to energy deprivation, and we discuss the consequences for metastatic escape. Understanding the energy requirements of amoeboid and other dissemination strategies offers rationales for improving therapeutic targeting of metastatic cancer progression.
Collapse
Affiliation(s)
- Maria Parlani
- Department of Cell Biology, Radboud University Medical Centre, Nijmegen 6525GA, The Netherlands
| | - Carolina Jorgez
- David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Peter Friedl
- Department of Cell Biology, Radboud University Medical Centre, Nijmegen 6525GA, The Netherlands; David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Cancer Genomics Center, 3584 CG Utrecht, The Netherlands.
| |
Collapse
|
7
|
Raudenská M, Petrláková K, Juriňáková T, Leischner Fialová J, Fojtů M, Jakubek M, Rösel D, Brábek J, Masařík M. Engine shutdown: migrastatic strategies and prevention of metastases. Trends Cancer 2023; 9:293-308. [PMID: 36804341 DOI: 10.1016/j.trecan.2023.01.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/29/2022] [Accepted: 01/03/2023] [Indexed: 02/17/2023]
Abstract
Most cancer-related deaths among patients with solid tumors are caused by metastases. Migrastatic strategies represent a unique therapeutic approach to prevent all forms of cancer cell migration and invasion. Because the migration machinery has been shown to promote metastatic dissemination, successful migrastatic therapy may reduce the need for high-dose cytotoxic therapies that are currently used to prevent the risk of metastatic dissemination. In this review we focus on anti-invasive and antimetastatic strategies that hold promise for the treatment of solid tumors. The best targets for migrastatic therapy would be those that are required by all forms of motility, such as ATP availability, mitochondrial metabolism, and cytoskeletal dynamics and cell contractility.
Collapse
Affiliation(s)
- Martina Raudenská
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, CZ-625 00 Brno, Czech Republic; Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, CZ-625 00 Brno, Czech Republic
| | - Kateřina Petrláková
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, CZ-625 00 Brno, Czech Republic; Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, CZ-625 00 Brno, Czech Republic
| | - Tamara Juriňáková
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, CZ-625 00 Brno, Czech Republic; Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, CZ-625 00 Brno, Czech Republic
| | - Jindřiška Leischner Fialová
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, CZ-625 00 Brno, Czech Republic; Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, CZ-625 00 Brno, Czech Republic
| | - Michaela Fojtů
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, CZ-625 00 Brno, Czech Republic; Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, CZ-625 00 Brno, Czech Republic
| | - Milan Jakubek
- BIOCEV (Biotechnology and Biomedicine Center in Vestec), First Faculty of Medicine, Charles University, Prumyslova 595, CZ-252 50 Vestec, Czech Republic
| | - Daniel Rösel
- Department of Cell Biology, BIOCEV, Faculty of Science, Charles University, CZ-252 50, Vestec, Prague-West, Czech Republic
| | - Jan Brábek
- Department of Cell Biology, BIOCEV, Faculty of Science, Charles University, CZ-252 50, Vestec, Prague-West, Czech Republic
| | - Michal Masařík
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, CZ-625 00 Brno, Czech Republic; Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, CZ-625 00 Brno, Czech Republic; BIOCEV (Biotechnology and Biomedicine Center in Vestec), First Faculty of Medicine, Charles University, Prumyslova 595, CZ-252 50 Vestec, Czech Republic.
| |
Collapse
|
8
|
Yang Z, Yan C, Ma J, Peng P, Ren X, Cai S, Shen X, Wu Y, Zhang S, Wang X, Qiu S, Zhou J, Fan J, Huang H, Gao Q. Lactylome analysis suggests lactylation-dependent mechanisms of metabolic adaptation in hepatocellular carcinoma. Nat Metab 2023; 5:61-79. [PMID: 36593272 DOI: 10.1038/s42255-022-00710-w] [Citation(s) in RCA: 248] [Impact Index Per Article: 124.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 11/11/2022] [Indexed: 01/03/2023]
Abstract
Enhanced glycolysis and accumulation of lactate is a common feature in various types of cancer. Intracellular lactate drives a recently described type of posttranslational modification, lysine lactylation (Kla), on core histones. However, the impact of lactylation on biological processes of tumour cells remains largely unknown. Here we show a global lactylome profiling on a prospectively collected hepatitis B virus-related hepatocellular carcinoma (HCC) cohort. Integrative lactylome and proteome analysis of the tumours and adjacent livers identifies 9,275 Kla sites, with 9,256 sites on non-histone proteins, indicating that Kla is a prevalent modification beyond histone proteins and transcriptional regulation. Notably, Kla preferentially affects enzymes involved in metabolic pathways, including the tricarboxylic acid cycle, and carbohydrate, amino acid, fatty acid and nucleotide metabolism. We further verify that lactylation at K28 inhibits the function of adenylate kinase 2, facilitating the proliferation and metastasis of HCC cells. Our study therefore reveals that Kla plays an important role in regulating cellular metabolism and may contribute to HCC progression.
Collapse
Affiliation(s)
- Zijian Yang
- Department of Liver Surgery and Transplantation, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Cong Yan
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jiaqiang Ma
- Department of Liver Surgery and Transplantation, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Panpan Peng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xuelian Ren
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shangli Cai
- Medical Department, Burning Rock Biotech, Guangdong, China
| | - Xia Shen
- Center for Tumor Diagnosis and Therapy, Jinshan Hospital, Fudan University, Shanghai, China
| | - Yingcheng Wu
- Department of Liver Surgery and Transplantation, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shu Zhang
- Department of Liver Surgery and Transplantation, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- Center for Tumor Diagnosis and Therapy, Jinshan Hospital, Fudan University, Shanghai, China
| | - Xiaoying Wang
- Department of Liver Surgery and Transplantation, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shuangjian Qiu
- Department of Liver Surgery and Transplantation, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jian Zhou
- Department of Liver Surgery and Transplantation, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jia Fan
- Department of Liver Surgery and Transplantation, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - He Huang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
| | - Qiang Gao
- Department of Liver Surgery and Transplantation, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China.
- Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
- State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China.
| |
Collapse
|
9
|
Krutilina RI, Playa H, Brooks DL, Schwab LP, Parke DN, Oluwalana D, Layman DR, Fan M, Johnson DL, Yue J, Smallwood H, Seagroves TN. HIF-Dependent CKB Expression Promotes Breast Cancer Metastasis, Whereas Cyclocreatine Therapy Impairs Cellular Invasion and Improves Chemotherapy Efficacy. Cancers (Basel) 2021; 14:cancers14010027. [PMID: 35008190 PMCID: PMC8749968 DOI: 10.3390/cancers14010027] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 12/16/2021] [Accepted: 12/19/2021] [Indexed: 01/14/2023] Open
Abstract
Simple Summary Targeting dysregulated cellular metabolism is a promising avenue to treat metastatic disease. The aim of our study was to identify genes downstream of the hypoxia-inducible factor (HIF)-1 transcription factor that are amenable to therapeutic intervention to treat metastatic breast cancer (MBC). We identified creatine kinase, brain isoform (CKB) as an HIF-dependent gene that strongly promotes invasion and metastasis in estrogen-receptor (ER)-negative breast cancer models. Deletion of Ckb also repressed glycolysis and mitochondrial respiration, leading to a reduction in intracellular ATP. Either the deletion of Ckb or inhibition of creatine kinase (CK) activity using the creatine analog cyclocreatine (cCr) repressed cell invasion, the formation of invadopodia and lung metastasis. In addition, when paired with paclitaxel or doxorubicin, cCr enhanced growth inhibition in an additive or synergistic manner. cCr may be an effective anti-metastatic agent in ER-negative, HIF-1α-positive breast cancers, targeting both cellular metabolism and motility, particularly when paired with conventional cytotoxic agents. Abstract The oxygen-responsive hypoxia inducible factor (HIF)-1 promotes several steps of the metastatic cascade. A hypoxic gene signature is enriched in triple-negative breast cancers (TNBCs) and is correlated with poor patient survival. Inhibiting the HIF transcription factors with small molecules is challenging; therefore, we sought to identify genes downstream of HIF-1 that could be targeted to block invasion and metastasis. Creatine kinase brain isoform (CKB) was identified as a highly differentially expressed gene in a screen of HIF-1 wild type and knockout mammary tumor cells derived from a transgenic model of metastatic breast cancer. CKB is a cytosolic enzyme that reversibly catalyzes the phosphorylation of creatine, generating phosphocreatine (PCr) in the forward reaction, and regenerating ATP in the reverse reaction. Creatine kinase activity is inhibited by the creatine analog cyclocreatine (cCr). Loss- and gain-of-function genetic approaches were used in combination with cCr therapy to define the contribution of CKB expression or creatine kinase activity to cell proliferation, migration, invasion, and metastasis in ER-negative breast cancers. CKB was necessary for cell invasion in vitro and strongly promoted tumor growth and lung metastasis in vivo. Similarly, cyclocreatine therapy repressed cell migration, cell invasion, the formation of invadopodia and lung metastasis. Moreover, in common TNBC cell line models, the addition of cCr to conventional cytotoxic chemotherapy agents was either additive or synergistic to repress tumor cell growth.
Collapse
Affiliation(s)
- Raisa I. Krutilina
- Center for Cancer Research, Department of Pathology and Laboratory Medicine, College of Medicine, The University of Tennessee Health Science Center, Cancer Research Building, 19 South Manassas Street, Memphis, TN 38163, USA; (R.I.K.); (H.P.); (D.L.B.); (L.P.S.); (D.N.P.); (D.O.); (M.F.); (J.Y.)
| | - Hilaire Playa
- Center for Cancer Research, Department of Pathology and Laboratory Medicine, College of Medicine, The University of Tennessee Health Science Center, Cancer Research Building, 19 South Manassas Street, Memphis, TN 38163, USA; (R.I.K.); (H.P.); (D.L.B.); (L.P.S.); (D.N.P.); (D.O.); (M.F.); (J.Y.)
| | - Danielle L. Brooks
- Center for Cancer Research, Department of Pathology and Laboratory Medicine, College of Medicine, The University of Tennessee Health Science Center, Cancer Research Building, 19 South Manassas Street, Memphis, TN 38163, USA; (R.I.K.); (H.P.); (D.L.B.); (L.P.S.); (D.N.P.); (D.O.); (M.F.); (J.Y.)
| | - Luciana P. Schwab
- Center for Cancer Research, Department of Pathology and Laboratory Medicine, College of Medicine, The University of Tennessee Health Science Center, Cancer Research Building, 19 South Manassas Street, Memphis, TN 38163, USA; (R.I.K.); (H.P.); (D.L.B.); (L.P.S.); (D.N.P.); (D.O.); (M.F.); (J.Y.)
| | - Deanna N. Parke
- Center for Cancer Research, Department of Pathology and Laboratory Medicine, College of Medicine, The University of Tennessee Health Science Center, Cancer Research Building, 19 South Manassas Street, Memphis, TN 38163, USA; (R.I.K.); (H.P.); (D.L.B.); (L.P.S.); (D.N.P.); (D.O.); (M.F.); (J.Y.)
| | - Damilola Oluwalana
- Center for Cancer Research, Department of Pathology and Laboratory Medicine, College of Medicine, The University of Tennessee Health Science Center, Cancer Research Building, 19 South Manassas Street, Memphis, TN 38163, USA; (R.I.K.); (H.P.); (D.L.B.); (L.P.S.); (D.N.P.); (D.O.); (M.F.); (J.Y.)
| | - Douglas R. Layman
- School of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163, USA;
| | - Meiyun Fan
- Center for Cancer Research, Department of Pathology and Laboratory Medicine, College of Medicine, The University of Tennessee Health Science Center, Cancer Research Building, 19 South Manassas Street, Memphis, TN 38163, USA; (R.I.K.); (H.P.); (D.L.B.); (L.P.S.); (D.N.P.); (D.O.); (M.F.); (J.Y.)
| | - Daniel L. Johnson
- Molecular Bioinformatics Core, Office of Research, The University of Tennessee Health Science Center, 71 South Manassas Street, Memphis, TN 38163, USA;
| | - Junming Yue
- Center for Cancer Research, Department of Pathology and Laboratory Medicine, College of Medicine, The University of Tennessee Health Science Center, Cancer Research Building, 19 South Manassas Street, Memphis, TN 38163, USA; (R.I.K.); (H.P.); (D.L.B.); (L.P.S.); (D.N.P.); (D.O.); (M.F.); (J.Y.)
| | - Heather Smallwood
- Department of Pediatrics, College of Medicine, The University of Tennessee Health Science Center, 71 South Manassas Street, Memphis, TN 38163, USA;
| | - Tiffany N. Seagroves
- Center for Cancer Research, Department of Pathology and Laboratory Medicine, College of Medicine, The University of Tennessee Health Science Center, Cancer Research Building, 19 South Manassas Street, Memphis, TN 38163, USA; (R.I.K.); (H.P.); (D.L.B.); (L.P.S.); (D.N.P.); (D.O.); (M.F.); (J.Y.)
- Correspondence: ; Tel.: +1-901-448-5018
| |
Collapse
|
10
|
Effects of an electric field on sleep quality and life span mediated by ultraviolet (UV)-A/blue light photoreceptor CRYPTOCHROME in Drosophila. Sci Rep 2021; 11:20543. [PMID: 34654874 PMCID: PMC8519966 DOI: 10.1038/s41598-021-99753-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 09/30/2021] [Indexed: 11/24/2022] Open
Abstract
Although electric fields (EF) exert beneficial effects on animal wound healing, differentiation, cancers and rheumatoid arthritis, the molecular mechanisms of these effects have remained unclear about a half century. Therefore, we aimed to elucidate the molecular mechanisms underlying EF effects in Drosophila melanogaster as a genetic animal model. Here we show that the sleep quality of wild type (WT) flies was improved by exposure to a 50-Hz (35 kV/m) constant electric field during the day time, but not during the night time. The effect was undetectable in cryptochrome mutant (cryb) flies. Exposure to a 50-Hz electric field under low nutrient conditions elongated the lifespan of male and female WT flies by ~ 18%, but not of several cry mutants and cry RNAi strains. Metabolome analysis indicated that the adenosine triphosphate (ATP) content was higher in intact WT than cry gene mutant strains exposed to an electric field. A putative magnetoreceptor protein and UV-A/blue light photoreceptor, CRYPTOCHROME (CRY) is involved in electric field (EF) receptors in animals. The present findings constitute hitherto unknown genetic evidence of a CRY-based system that is electric field sensitive in animals.
Collapse
|
11
|
Li J, Fang Y, Wu D. Mechanical forces and metabolic changes cooperate to drive cellular memory and endothelial phenotypes. CURRENT TOPICS IN MEMBRANES 2021; 87:199-253. [PMID: 34696886 PMCID: PMC8639155 DOI: 10.1016/bs.ctm.2021.07.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Endothelial cells line the innermost layer of arterial, venous, and lymphatic vascular tree and accordingly are subject to hemodynamic, stretch, and stiffness mechanical forces. Normally quiescent, endothelial cells have a hemodynamic set point and become "activated" in response to disturbed hemodynamics, which may signal impending nutrient or gas depletion. Endothelial cells in the majority of tissue beds are normally inactivated and maintain vessel barrier functions, are anti-inflammatory, anti-coagulant, and anti-thrombotic. However, under aberrant mechanical forces, endothelial signaling transforms in response, resulting cellular changes that herald pathological diseases. Endothelial cell metabolism is now recognized as the primary intermediate pathway that undergirds cellular transformation. In this review, we discuss the various mechanical forces endothelial cells sense in the large vessels, microvasculature, and lymphatics, and how changes in environmental mechanical forces result in changes in metabolism, which ultimately influence cell physiology, cellular memory, and ultimately disease initiation and progression.
Collapse
Affiliation(s)
- Jin Li
- Committee on Molecular Metabolism and Nutrition, Biological Sciences Division, University of Chicago, Chicago, IL, United States; Department of Medicine, Biological Sciences Division, University of Chicago, Chicago, IL, United States
| | - Yun Fang
- Committee on Molecular Metabolism and Nutrition, Biological Sciences Division, University of Chicago, Chicago, IL, United States; Department of Medicine, Biological Sciences Division, University of Chicago, Chicago, IL, United States
| | - David Wu
- Committee on Molecular Metabolism and Nutrition, Biological Sciences Division, University of Chicago, Chicago, IL, United States; Department of Medicine, Biological Sciences Division, University of Chicago, Chicago, IL, United States.
| |
Collapse
|
12
|
Lacerda-Abreu MA, Russo-Abrahão T, Rocco-Machado N, Cosentino-Gomes D, Dick CF, Carvalho-Kelly LF, Cunha Nascimento MT, Rocha-Vieira TC, Meyer-Fernandes JR. Hydrogen Peroxide Generation as an Underlying Response to High Extracellular Inorganic Phosphate (Pi) in Breast Cancer Cells. Int J Mol Sci 2021; 22:ijms221810096. [PMID: 34576256 PMCID: PMC8468810 DOI: 10.3390/ijms221810096] [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: 04/23/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 11/16/2022] Open
Abstract
According to the growth rate hypothesis (GRH), tumour cells have high inorganic phosphate (Pi) demands due to accelerated proliferation. Compared to healthy individuals, cancer patients present with a nearly 2.5-fold higher Pi serum concentration. In this work, we show that an increasing concentration of Pi had the opposite effect on Pi-transporters only in MDA-MB-231 when compared to other breast cell lines: MCF-7 or MCF10-A (non-tumoural breast cell line). Here, we show for the first time that high extracellular Pi concentration mediates ROS production in TNBC (MDA-MB-231). After a short-time exposure (1 h), Pi hyperpolarizes the mitochondrial membrane, increases mitochondrial ROS generation, impairs oxygen (O2) consumption and increases PKC activity. However, after 24 h Pi-exposure, the source of H2O2 seems to shift from mitochondria to an NADPH oxidase enzyme (NOX), through activation of PKC by H2O2. Exogenous-added H2O2 modulated Pi-transporters the same way as extracellular high Pi, which could be reversed by the addition of the antioxidant N-acetylcysteine (NAC). NAC was also able to abolish Pi-induced Epithelial-mesenchymal transition (EMT), migration and adhesion of MDA-MB-231. We believe that Pi transporters support part of the energy required for the metastatic processes stimulated by Pi and trigger Pi-induced H2O2 production as a signalling response to promote cell migration and adhesion.
Collapse
Affiliation(s)
- Marco Antonio Lacerda-Abreu
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, RJ, Brazil; (M.A.L.-A.); (T.R.-A.); (N.R.-M.); (D.C.-G.); (C.F.D.); (L.F.C.-K.); (M.T.C.N.); (T.C.R.-V.)
- Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Bioimagem, Rio de Janeiro 21941-590, RJ, Brazil
| | - Thais Russo-Abrahão
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, RJ, Brazil; (M.A.L.-A.); (T.R.-A.); (N.R.-M.); (D.C.-G.); (C.F.D.); (L.F.C.-K.); (M.T.C.N.); (T.C.R.-V.)
- Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Bioimagem, Rio de Janeiro 21941-590, RJ, Brazil
| | - Nathália Rocco-Machado
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, RJ, Brazil; (M.A.L.-A.); (T.R.-A.); (N.R.-M.); (D.C.-G.); (C.F.D.); (L.F.C.-K.); (M.T.C.N.); (T.C.R.-V.)
- Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Bioimagem, Rio de Janeiro 21941-590, RJ, Brazil
- National Heart, Lung, and Blood Institute, NIH, Bethesda, Rockville, MD 20814, USA
| | - Daniela Cosentino-Gomes
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, RJ, Brazil; (M.A.L.-A.); (T.R.-A.); (N.R.-M.); (D.C.-G.); (C.F.D.); (L.F.C.-K.); (M.T.C.N.); (T.C.R.-V.)
- Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Bioimagem, Rio de Janeiro 21941-590, RJ, Brazil
- Departamento de Bioquímica, Universidade Federal Rural do Rio de Janeiro, Seropédica 23890-000, RJ, Brazil
| | - Claudia Fernanda Dick
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, RJ, Brazil; (M.A.L.-A.); (T.R.-A.); (N.R.-M.); (D.C.-G.); (C.F.D.); (L.F.C.-K.); (M.T.C.N.); (T.C.R.-V.)
- Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Bioimagem, Rio de Janeiro 21941-590, RJ, Brazil
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, RJ, Brazil
| | - Luiz Fernando Carvalho-Kelly
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, RJ, Brazil; (M.A.L.-A.); (T.R.-A.); (N.R.-M.); (D.C.-G.); (C.F.D.); (L.F.C.-K.); (M.T.C.N.); (T.C.R.-V.)
- Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Bioimagem, Rio de Janeiro 21941-590, RJ, Brazil
| | - Michelle Tanny Cunha Nascimento
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, RJ, Brazil; (M.A.L.-A.); (T.R.-A.); (N.R.-M.); (D.C.-G.); (C.F.D.); (L.F.C.-K.); (M.T.C.N.); (T.C.R.-V.)
- Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Bioimagem, Rio de Janeiro 21941-590, RJ, Brazil
| | - Thaís Cristino Rocha-Vieira
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, RJ, Brazil; (M.A.L.-A.); (T.R.-A.); (N.R.-M.); (D.C.-G.); (C.F.D.); (L.F.C.-K.); (M.T.C.N.); (T.C.R.-V.)
- Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Bioimagem, Rio de Janeiro 21941-590, RJ, Brazil
| | - José Roberto Meyer-Fernandes
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, RJ, Brazil; (M.A.L.-A.); (T.R.-A.); (N.R.-M.); (D.C.-G.); (C.F.D.); (L.F.C.-K.); (M.T.C.N.); (T.C.R.-V.)
- Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Bioimagem, Rio de Janeiro 21941-590, RJ, Brazil
- Correspondence: ; Tel.: +55-21-3938-6781; Fax: +55-21-2270-8647
| |
Collapse
|
13
|
Bahadoran A, Bezavada L, Smallwood HS. Fueling influenza and the immune response: Implications for metabolic reprogramming during influenza infection and immunometabolism. Immunol Rev 2021; 295:140-166. [PMID: 32320072 DOI: 10.1111/imr.12851] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/19/2020] [Accepted: 02/24/2020] [Indexed: 12/11/2022]
Abstract
Recent studies support the notion that glycolysis and oxidative phosphorylation are rheostats in immune cells whose bioenergetics have functional outputs in terms of their biology. Specific intrinsic and extrinsic molecular factors function as molecular potentiometers to adjust and control glycolytic to respiratory power output. In many cases, these potentiometers are used by influenza viruses and immune cells to support pathogenesis and the host immune response, respectively. Influenza virus infects the respiratory tract, providing a specific environmental niche, while immune cells encounter variable nutrient concentrations as they migrate in response to infection. Immune cell subsets have distinct metabolic programs that adjust to meet energetic and biosynthetic requirements to support effector functions, differentiation, and longevity in their ever-changing microenvironments. This review details how influenza coopts the host cell for metabolic reprogramming and describes the overlap of these regulatory controls in immune cells whose function and fate are dictated by metabolism. These details are contextualized with emerging evidence of the consequences of influenza-induced changes in metabolic homeostasis on disease progression.
Collapse
Affiliation(s)
- Azadeh Bahadoran
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Lavanya Bezavada
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Heather S Smallwood
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
14
|
Wu Y, Zanotelli MR, Zhang J, Reinhart-King CA. Matrix-driven changes in metabolism support cytoskeletal activity to promote cell migration. Biophys J 2021; 120:1705-1717. [PMID: 33705759 PMCID: PMC8204337 DOI: 10.1016/j.bpj.2021.02.044] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 02/03/2021] [Accepted: 02/23/2021] [Indexed: 01/21/2023] Open
Abstract
The microenvironment provides both active and passive mechanical cues that regulate cell morphology, adhesion, migration, and metabolism. Although the cellular response to those mechanical cues often requires energy-intensive actin cytoskeletal remodeling and actomyosin contractility, it remains unclear how cells dynamically adapt their metabolic activity to altered mechanical cues to support migration. Here, we investigated the changes in cellular metabolic activity in response to different two-dimensional and three-dimensional microenvironmental conditions and how these changes relate to cytoskeletal activity and migration. Utilizing collagen micropatterning on polyacrylamide gels, intracellular energy levels and oxidative phosphorylation were found to be correlated with cell elongation and spreading and necessary for membrane ruffling. To determine whether this relationship holds in more physiological three-dimensional matrices, collagen matrices were used to show that intracellular energy state was also correlated with protrusive activity and increased with matrix density. Pharmacological inhibition of oxidative phosphorylation revealed that cancer cells rely on oxidative phosphorylation to meet the elevated energy requirements for protrusive activity and migration in denser matrices. Together, these findings suggest that mechanical regulation of cytoskeletal activity during spreading and migration by the physical microenvironment is driven by an altered metabolic profile.
Collapse
Affiliation(s)
- Yusheng Wu
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Matthew R Zanotelli
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee; Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York
| | - Jian Zhang
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | | |
Collapse
|
15
|
Romani P, Valcarcel-Jimenez L, Frezza C, Dupont S. Crosstalk between mechanotransduction and metabolism. Nat Rev Mol Cell Biol 2021; 22:22-38. [PMID: 33188273 DOI: 10.1038/s41580-020-00306-w] [Citation(s) in RCA: 250] [Impact Index Per Article: 62.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2020] [Indexed: 12/22/2022]
Abstract
Mechanical forces shape cells and tissues during development and adult homeostasis. In addition, they also signal to cells via mechanotransduction pathways to control cell proliferation, differentiation and death. These processes require metabolism of nutrients for both energy generation and biosynthesis of macromolecules. However, how cellular mechanics and metabolism are connected is still poorly understood. Here, we discuss recent evidence indicating how the mechanical cues exerted by the extracellular matrix (ECM), cell-ECM and cell-cell adhesion complexes influence metabolic pathways. Moreover, we explore the energy and metabolic requirements associated with cell mechanics and ECM remodelling, implicating a reciprocal crosstalk between cell mechanics and metabolism.
Collapse
Affiliation(s)
- Patrizia Romani
- Department of Molecular Medicine, University of Padua Medical School, Padua, Italy
| | | | - Christian Frezza
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge, UK.
| | - Sirio Dupont
- Department of Molecular Medicine, University of Padua Medical School, Padua, Italy.
| |
Collapse
|
16
|
Wolfe K, Kamata R, Coutinho K, Inoue T, Sasaki AT. Metabolic Compartmentalization at the Leading Edge of Metastatic Cancer Cells. Front Oncol 2020; 10:554272. [PMID: 33224873 PMCID: PMC7667250 DOI: 10.3389/fonc.2020.554272] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 09/29/2020] [Indexed: 12/30/2022] Open
Abstract
Despite advances in targeted therapeutics and understanding in molecular mechanisms, metastasis remains a substantial obstacle for cancer treatment. Acquired genetic mutations and transcriptional changes can promote the spread of primary tumor cells to distant tissues. Additionally, recent studies have uncovered that metabolic reprogramming of cancer cells is tightly associated with cancer metastasis. However, whether intracellular metabolism is spatially and temporally regulated for cancer cell migration and invasion is understudied. In this review, we highlight the emergence of a concept, termed “membraneless metabolic compartmentalization,” as one of the critical mechanisms that determines the metastatic capacity of cancer cells. In particular, we focus on the compartmentalization of purine nucleotide metabolism (e.g., ATP and GTP) at the leading edge of migrating cancer cells through the uniquely phase-separated microdomains where dynamic exchange of nucleotide metabolic enzymes takes place. We will discuss how future insights may usher in a novel class of therapeutics specifically targeting the metabolic compartmentalization that drives tumor metastasis.
Collapse
Affiliation(s)
- Kara Wolfe
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Department of Cancer Biology, University of Cincinnati College of Medicine, OH, United States
| | - Ryo Kamata
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan.,Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| | - Kester Coutinho
- Department of Cell Biology and Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Takanari Inoue
- Department of Cell Biology and Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Atsuo T Sasaki
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Department of Cancer Biology, University of Cincinnati College of Medicine, OH, United States.,Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan.,Department of Neurosurgery, Brain Tumor Center at UC Gardner Neuroscience Institute, Cincinnati, OH, United States
| |
Collapse
|
17
|
Sneeggen M, Guadagno NA, Progida C. Intracellular Transport in Cancer Metabolic Reprogramming. Front Cell Dev Biol 2020; 8:597608. [PMID: 33195279 PMCID: PMC7661548 DOI: 10.3389/fcell.2020.597608] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 10/12/2020] [Indexed: 12/12/2022] Open
Abstract
Tumor progression is a complex process consisting of several steps characterized by alterations in cellular behavior and morphology. These steps include uncontrolled cell division and proliferation, invasiveness and metastatic ability. Throughout these phases, cancer cells encounter a changing environment and a variety of metabolic stress. To meet their needs for energy while they proliferate and survive in their new environment, tumor cells need to continuously fine-tune their metabolism. The connection between intracellular transport and metabolic reprogramming during cancer progression is emerging as a central process of cellular adaptation to these changes. The trafficking of proteolytic enzymes, surface receptors, but also the regulation of downstream pathways, are all central to cancer progression. In this review, we summarize different hallmarks of cancer with a special focus on the role of intracellular trafficking in cell proliferation, epithelial to mesenchymal transition as well as invasion. We will further emphasize how intracellular trafficking contributes to the regulation of energy consumption and metabolism during these steps of cancer progression.
Collapse
Affiliation(s)
- Marte Sneeggen
- Department of Biosciences, University of Oslo, Oslo, Norway
| | | | - Cinzia Progida
- Department of Biosciences, University of Oslo, Oslo, Norway
| |
Collapse
|
18
|
Rezinciuc S, Bezavada L, Bahadoran A, Duan S, Wang R, Lopez-Ferrer D, Finkelstein D, McGargill MA, Green DR, Pasa-Tolic L, Smallwood HS. Dynamic metabolic reprogramming in dendritic cells: An early response to influenza infection that is essential for effector function. PLoS Pathog 2020; 16:e1008957. [PMID: 33104753 PMCID: PMC7707590 DOI: 10.1371/journal.ppat.1008957] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 12/01/2020] [Accepted: 09/03/2020] [Indexed: 01/19/2023] Open
Abstract
Infection with the influenza virus triggers an innate immune response that initiates the adaptive response to halt viral replication and spread. However, the metabolic response fueling the molecular mechanisms underlying changes in innate immune cell homeostasis remain undefined. Although influenza increases parasitized cell metabolism, it does not productively replicate in dendritic cells. To dissect these mechanisms, we compared the metabolism of dendritic cells to that of those infected with active and inactive influenza A virus and those treated with toll-like receptor agonists. Using quantitative mass spectrometry, pulse chase substrate utilization assays and metabolic flux measurements, we found global metabolic changes in dendritic cells 17 hours post infection, including significant changes in carbon commitment via glycolysis and glutaminolysis, as well as mitochondrial respiration. Influenza infection of dendritic cells led to a metabolic phenotype distinct from that induced by TLR agonists, with significant resilience in terms of metabolic plasticity. We identified c-Myc as one transcription factor modulating this response. Restriction of c-Myc activity or mitochondrial substrates significantly changed the immune functions of dendritic cells, such as reducing motility and T cell activation. Transcriptome analysis of inflammatory dendritic cells isolated following influenza infection showed similar metabolic reprogramming occurs in vivo. Thus, early in the infection process, dendritic cells respond with global metabolic restructuring, that is present in inflammatory lung dendritic cells after infection, and this is important for effector function. These findings suggest metabolic switching in dendritic cells plays a vital role in initiating the immune response to influenza infection. Dendritic cells are critical in mounting an effective immune response to influenza infection by initiating the immune response to influenza and activating the adaptive response to mediate viral clearance and manifest immune memory for protection against subsequent infections. We found dendritic cells undergo a profound metabolic shift after infection. They alter the concentration and location of hundreds of proteins, including c-Myc, facilitating a shift to a highly glycolytic phenotype that is also flexible in terms of fueling respiration. Nonetheless, we found limiting access to specific metabolic pathways or substrates diminished key immune functions. We previously described an immediate, fixed hypermetabolic state in infected respiratory epithelial cells. Here we present data indicating the metabolic response of dendritic cells is increased yet flexible, distinct from what we previously showed for epithelial cells. Additionally, we demonstrate dendritic cells tailor their metabolic response to the pathogen or TLR stimulus. This metabolic reprogramming occurs rapidly in vitro and is sustained in inflammatory dendritic cells in vivo for at least 9 days following influenza infection. These studies introduce the possibility of modulating the immune response to viral infection using customized metabolic therapy to enhance or diminish the function of specific cells.
Collapse
Affiliation(s)
- Svetlana Rezinciuc
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Lavanya Bezavada
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Azadeh Bahadoran
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Susu Duan
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Ruoning Wang
- Center for Childhood Cancer and Blood Disease, The Research Institute at Nationwide Children's Hospital, The Ohio State University School of Medicine, Columbus, Ohio, United States of America
| | - Daniel Lopez-Ferrer
- Chromatography and Mass Spectrometry Division, Thermo Fisher Scientific, CA, United States of America
| | - David Finkelstein
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Maureen A. McGargill
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Douglas R. Green
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Ljiljana Pasa-Tolic
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington, United States of America
| | - Heather S. Smallwood
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
19
|
Bressan C, Pecora A, Gagnon D, Snapyan M, Labrecque S, De Koninck P, Parent M, Saghatelyan A. The dynamic interplay between ATP/ADP levels and autophagy sustain neuronal migration in vivo. eLife 2020; 9:56006. [PMID: 32985978 PMCID: PMC7556871 DOI: 10.7554/elife.56006] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 09/26/2020] [Indexed: 02/07/2023] Open
Abstract
Cell migration is a dynamic process that entails extensive protein synthesis and recycling, structural remodeling, and considerable bioenergetic demand. Autophagy is one of the pathways that maintain cellular homeostasis. Time-lapse imaging of autophagosomes and ATP/ADP levels in migrating cells in the rostral migratory stream of mouse revealed that decreases in ATP levels force cells into the stationary phase and induce autophagy. Pharmacological or genetic impairments of autophagy in neuroblasts using either bafilomycin, inducible conditional mice, or CRISPR/Cas9 gene editing decreased cell migration due to the longer duration of the stationary phase. Autophagy is modulated in response to migration-promoting and inhibiting molecular cues and is required for the recycling of focal adhesions. Our results show that autophagy and energy consumption act in concert in migrating cells to dynamically regulate the pace and periodicity of the migratory and stationary phases to sustain neuronal migration.
Collapse
Affiliation(s)
- Cedric Bressan
- CERVO Brain Research Center, Quebec City, Canada.,Université Laval, Quebec City, Canada
| | - Alessandra Pecora
- CERVO Brain Research Center, Quebec City, Canada.,Université Laval, Quebec City, Canada
| | - Dave Gagnon
- CERVO Brain Research Center, Quebec City, Canada.,Université Laval, Quebec City, Canada
| | - Marina Snapyan
- CERVO Brain Research Center, Quebec City, Canada.,Université Laval, Quebec City, Canada
| | - Simon Labrecque
- CERVO Brain Research Center, Quebec City, Canada.,Université Laval, Quebec City, Canada
| | - Paul De Koninck
- CERVO Brain Research Center, Quebec City, Canada.,Université Laval, Quebec City, Canada
| | - Martin Parent
- CERVO Brain Research Center, Quebec City, Canada.,Université Laval, Quebec City, Canada
| | - Armen Saghatelyan
- CERVO Brain Research Center, Quebec City, Canada.,Université Laval, Quebec City, Canada
| |
Collapse
|
20
|
Dumas SJ, García-Caballero M, Carmeliet P. Metabolic Signatures of Distinct Endothelial Phenotypes. Trends Endocrinol Metab 2020; 31:580-595. [PMID: 32622584 DOI: 10.1016/j.tem.2020.05.009] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 05/08/2020] [Accepted: 05/26/2020] [Indexed: 02/08/2023]
Abstract
Angiogenesis is crucial for the development of the blood vasculature during embryogenesis, but also contributes to cancer and other diseases. While therapeutic targeting of endothelial cells (ECs) through growth factor inhibition is limited by insufficient efficacy and resistance, a new paradigm for modulating angiogenesis by targeting EC metabolism has emerged. Findings from the past decade highlight how ECs adapt their metabolism to proliferate or migrate during vessel sprouting, or to maintain the vascular barrier and protect themselves against oxidative stress in the high-oxygen environment they are exposed to in healthy conditions. We overview key endothelial metabolic pathways underlying the different EC phenotypes, as well as potential opportunities for targeting EC metabolism in therapeutic settings.
Collapse
Affiliation(s)
- Sébastien J Dumas
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, Katholieke Universiteit (KU) Leuven, Leuven, B 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Vlaams Instituut voor Biotechnologie (VIB), Leuven, B 3000, Belgium
| | - Melissa García-Caballero
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, Katholieke Universiteit (KU) Leuven, Leuven, B 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Vlaams Instituut voor Biotechnologie (VIB), Leuven, B 3000, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, Katholieke Universiteit (KU) Leuven, Leuven, B 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Vlaams Instituut voor Biotechnologie (VIB), Leuven, B 3000, Belgium.
| |
Collapse
|
21
|
McColloch A, Liebman C, Liu H, Cho M. Alterted Adipogenesis of Human Mesenchymal Stem Cells by Photobiomodulation Using 1064 nm Laser Light. Lasers Surg Med 2020; 53:263-274. [PMID: 32495397 DOI: 10.1002/lsm.23278] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/27/2020] [Accepted: 05/23/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND OBJECTIVES Photobiomodulation (PBM) describes the influence of light irradiation on biological tissues. Laser light in the near-infrared (NIR) spectrum has been shown to mitigate pain, reduce inflammation, and promote wound healing. The cellular mechanism that mediates PBM's effects is generally accepted to be at the site of the mitochondria, leading to an increased flux through the electron transport chain and adenosine triphosphate (ATP) production. Moreover, PBM has been demonstrated to reduce oxidative stress through an increased production of reactive oxygen species (ROS)-sequestering enzymes. The aim of the study is to determine whether these PBM-induced effects expedite or interfere with the intended stem cell differentiation to the adipogenic lineage. STUDY DESIGN/MATERIALS AND METHODS To determine the effects of 1064 nm laser irradiation (fluence of 8.8-26.4 J/cm2 ) on human mesenchymal stem cells (hMSCs) undergoing adipogenic differentiation, the ATP and ROS levels, and adipogenic markers were quantitatively measured. RESULTS At a low fluence (8.8 J/cm2 ) the ATP increase was essentially negligible, whereas a higher fluence induced a significant increase. In the laser-stimulated cells, PBM over time decreased the ROS level compared with the non-treated control group and significantly reduced the extent of adipogenesis. A reduction in the ROS level was correlated with a diminished lipid accumulation, reduced production of adipose-specific genetic markers, and delayed the chemically intended adipogenesis. CONCLUSION We characterized the use of NIR light exposure to modulate adipogenesis. Both the ATP and ROS levels in hMSCs responded to different energy densities. The current study is expected to contribute significantly to the growing field of PBM as well as stem cell tissue engineering by demonstrating the wavelength-dependent responses of hMSC differentiation. Lasers Surg. Med. © 2020 Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Andrew McColloch
- Department of Bioengineering, The University of Texas at Arlington, Arlington, Texas, 76019
| | - Caleb Liebman
- Department of Bioengineering, The University of Texas at Arlington, Arlington, Texas, 76019
| | - Hanli Liu
- Department of Bioengineering, The University of Texas at Arlington, Arlington, Texas, 76019
| | - Michael Cho
- Department of Bioengineering, The University of Texas at Arlington, Arlington, Texas, 76019
| |
Collapse
|
22
|
Klepinin A, Zhang S, Klepinina L, Rebane-Klemm E, Terzic A, Kaambre T, Dzeja P. Adenylate Kinase and Metabolic Signaling in Cancer Cells. Front Oncol 2020; 10:660. [PMID: 32509571 PMCID: PMC7248387 DOI: 10.3389/fonc.2020.00660] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 04/08/2020] [Indexed: 12/23/2022] Open
Abstract
A hallmark of cancer cells is the ability to rewire their bioenergetics and metabolic signaling circuits to fuel their uncontrolled proliferation and metastasis. Adenylate kinase (AK) is the critical enzyme in the metabolic monitoring of cellular adenine nucleotide homeostasis. It also directs AK→ AMP→ AMPK signaling controlling cell cycle and proliferation, and ATP energy transfer from mitochondria to distribute energy among cellular processes. The significance of AK isoform network in the regulation of a variety of cellular processes, which include cell differentiation and motility, is rapidly growing. Adenylate kinase 2 (AK2) isoform, localized in intermembrane and intra-cristae space, is vital for mitochondria nucleotide exchange and ATP export. AK2 deficiency disrupts cell energetics, causes severe human diseases, and is embryonically lethal in mice, signifying the importance of catalyzed phosphotransfer in cellular energetics. Suppression of AK phosphotransfer and AMP generation in cancer cells and consequently signaling through AMPK could be an important factor in the initiation of cancerous transformation, unleashing uncontrolled cell cycle and growth. Evidence also builds up that shift in AK isoforms is used later by cancer cells for rewiring energy metabolism to support their high proliferation activity and tumor progression. As cell motility is an energy-consuming process, positioning of AK isoforms to increased energy consumption sites could be an essential factor to incline cancer cells to metastases. In this review, we summarize recent advances in studies of the significance of AK isoforms involved in cancer cell metabolism, metabolic signaling, metastatic potential, and a therapeutic target.
Collapse
Affiliation(s)
- Aleksandr Klepinin
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | - Song Zhang
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Ljudmila Klepinina
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | - Egle Rebane-Klemm
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | - Andre Terzic
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Tuuli Kaambre
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | - Petras Dzeja
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
23
|
Joshi B, Pawling J, Shankar J, Pacholczyk K, Kim Y, Tran W, Meng F, Rahman AMA, Foster LJ, Leong HS, Dennis JW, Nabi IR. Caveolin-1 Y14 phosphorylation suppresses tumor growth while promoting invasion. Oncotarget 2019; 10:6668-6677. [PMID: 31803361 PMCID: PMC6877104 DOI: 10.18632/oncotarget.27313] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 10/26/2019] [Indexed: 01/07/2023] Open
Abstract
Caveolin-1 is a transmembrane protein with both tumor promoter and suppressor functions that remain poorly understood. Cav1 phosphorylation by Src kinase on tyrosine 14 is closely associated with focal adhesion dynamics and tumor cell migration, however the role of pCav1 in vivo in tumor progression remains poorly characterized. Herein, we expressed phosphomimetic Y14D, wild type, and non-phosphorylatable Y14F forms of Cav1 in MDA-MB-435 cancer cells. Expression of Cav1Y14D reduced cell proliferation and induced the TP53 tumor suppressor. Ectopic expression in MDA-MB-435 cells of Y14 phosphorylatable Cav1 was required for induction of TP53 in response to oxidative stress. Cav1Y14D promotes an apparent reversal of the Warburg effect and markedly inhibited tumor growth in vivo. However, Cav1 induced pseudopodial recruitment of glycolytic enzymes, and time-lapse intravital imaging showed increased invadopodia protrusion and extravasation into blood vessels for Cav1WT and Y14D but not for Y14F. Our results suggest that Cav1 Y14 phosphorylation levels play a role in the conflicting demands on metabolic resources associated with cancer cell proliferation versus motility.
Collapse
Affiliation(s)
- Bharat Joshi
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Judy Pawling
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Jay Shankar
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Karina Pacholczyk
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Yohan Kim
- Translational Prostate Cancer Research Group, London Regional Cancer Program, University of Western Ontario, London, Canada
| | - Wynn Tran
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Fanrui Meng
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Anas M Abdel Rahman
- Department of Genetics, King Faisal Specialist Hospital and Research Centre (KFSHRC), Riyadh, Saudi Arabia
| | - Leonard J Foster
- Centre for High-throughput Biology, University of British Columbia, Vancouver, Canada
| | - Hon S Leong
- Translational Prostate Cancer Research Group, London Regional Cancer Program, University of Western Ontario, London, Canada
| | - James W Dennis
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Ivan R Nabi
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| |
Collapse
|
24
|
Energetic costs regulated by cell mechanics and confinement are predictive of migration path during decision-making. Nat Commun 2019; 10:4185. [PMID: 31519914 PMCID: PMC6744572 DOI: 10.1038/s41467-019-12155-z] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 08/15/2019] [Indexed: 02/07/2023] Open
Abstract
Cell migration during the invasion-metastasis cascade requires cancer cells to navigate a spatially complex microenvironment that presents directional choices to migrating cells. Here, we investigate cellular energetics during migration decision-making in confined spaces. Theoretical and experimental data show that energetic costs for migration through confined spaces are mediated by a balance between cell and matrix compliance as well as the degree of spatial confinement to direct decision-making. Energetic costs, driven by the cellular work needed to generate force for matrix displacement, increase with increasing cell stiffness, matrix stiffness, and degree of spatial confinement, limiting migration. By assessing energetic costs between possible migration paths, we can predict the probability of migration choice. Our findings indicate that motility in confined spaces imposes high energetic demands on migrating cells, and cells migrate in the direction of least confinement to minimize energetic costs. Therefore, therapeutically targeting metabolism may limit cancer cell migration and metastasis.
Collapse
|
25
|
A co-expressed gene status of adenylate kinase 1/4 reveals prognostic gene signature associated with prognosis and sensitivity to EGFR targeted therapy in lung adenocarcinoma. Sci Rep 2019; 9:12329. [PMID: 31444368 PMCID: PMC6707279 DOI: 10.1038/s41598-019-48243-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 08/01/2019] [Indexed: 02/06/2023] Open
Abstract
Cancer cells utilize altered bioenergetics to fuel uncontrolled proliferation and progression. At the core of bioenergetics, adenine nucleotides are the building blocks for nucleotide synthesis, energy transfer and diverse metabolic processes. Adenylate kinases (AK) are ubiquitous phosphotransferases that catalyze the conversion of adenine nucleotides and regulate the homeostasis of nucleotide ratios within cellular compartments. Recently, different isoforms of AK have been shown to induce metabolic reprograming in cancer and were identified as biomarkers for predicting disease progression. Here we aim to systemically analyze the impact of all AK-associated gene signatures on lung adenocarcinoma patient survival and decipher the value for therapeutic interventions. By analyzing TCGA Lung Adenocarcinoma (LUAD) RNA Seq data, we found gene signatures from AK4 and AK1 have higher percentage of prognostic genes compared to other AK-gene signatures. A 118-gene signature was identified from consensus gene expression in AK1 and AK4 prognostic gene signatures. Immunohistochemistry (IHC) analyses in 140 lung adenocarcinoma patients showed overexpression of AK4 significantly correlated with worse overall survival (P = 0.001) whereas overexpression of AK1 significantly associated with good prognosis (P = 0.009). Furthermore, reduced AK4 expression by shRNA reduced the EGFR protein expression in EGFR mutation cells. The inhibition of AK4-AK1 signal might provide a potential target for synergistic effect in target therapy in lung cancer patients.
Collapse
|
26
|
Abstract
Adenylate kinase is a small, usually monomeric, enzyme found in every living thing due to its crucial role in energetic metabolism. This paper outlines the most relevant data about adenylate kinases isoforms, and the connection between dysregulation or mutation of human adenylate kinase and medical conditions. The following datadases were consulted: National Centre for Biotechnology Information, Protein Data Bank, and Mouse Genomic Informatics. The SmartBLAST tool, EMBOSS Needle Program, and Clustal Omega Program were used to analyze the best protein match, and to perform pairwise sequence alignment and multiple sequence alignment. Human adenylate kinase genes are located on different chromosomes, six of them being on the chromosomes 1 and 9. The adenylate kinases' intracellular localization and organ distribution explain their dysregulation in many diseases. The cytosolic isoenzyme 1 and the mitochondrial isoenzyme 2 are the main adenylate kinases that are integrated in the vast network of inflammatory modulators. The cytosolic isoenzyme 5 is correlated with limbic encephalitis and Leu673Pro mutation of the isoenzyme 7 leads to primary male infertility due to impairment of the ciliary function. The impairment of the mitochondrial isoenzymes 2 and 4 is demonstrated in neuroblastoma or glioma. The adenylate kinases are disease modifier that can assess the risk of diseases where oxidative stress plays a crucial role in pathogenesis like metabolic syndrome or neurodegenerative diseases. Because adenylate kinases has ATP as substrate, they are integrated in the global network of energetic process of any organism therefore are valid target for new pharmaceutical compounds.
Collapse
Affiliation(s)
- Mihaela Ileana Ionescu
- Department of Microbiology, Faculty of Medicine, Iuliu Haţieganu University of Medicine and Pharmacy, 6 Louis Pasteur, Cluj-Napoca, 400349, Romania.
- County Emergency Clinical Hospital, Cluj-Napoca, Romania.
| |
Collapse
|
27
|
Zhao X, Lang L, He L, Gao L, Chyan D, Xiong Y, Li H, Peng H, Teng Y. Intracellular reduction in ATP levels contributes to CYT997-induced suppression of metastasis of head and neck squamous carcinoma. J Cell Mol Med 2019; 23:1174-1182. [PMID: 30450674 PMCID: PMC6349165 DOI: 10.1111/jcmm.14017] [Citation(s) in RCA: 6] [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: 02/09/2018] [Revised: 10/16/2018] [Accepted: 10/17/2018] [Indexed: 11/29/2022] Open
Abstract
The incidence rate of head and neck squamous cell carcinoma (HNSCC) has steadily increased over the past decade. However, treatment options for metastatic HNSCC are often limited and the 5-year survival rate has remained static. Therefore, the development and assessment of more efficient but less toxic therapeutic strategies is an unmet need for treatment of more extensive HNSCC. Here, we report that CYT997, a novel microtubule-disrupting agent, exerts strong activity in inhibiting HNSCC cell invasion and metastasis. The loss of invasion capacity by CYT997 was accompanied by an associated increase in cell adhesion and the reversal of epithelial-mesenchymal transition (EMT). Increased expression of E-cadherin protein and decreased expression of Vimentin protein became evident in HNSCC cells following CYT997 exposure, which were consistently observed in HNSCC xenografts from the mice receiving CYT997. Moreover, the capacity of invasive HNSCC cells to form pulmonary metastases was significantly blocked with CYT997 treatment, indicating that the diminishment of EMT traits contributes to CYT997-suppressed metastasis. Intriguingly, CYT997 impaired intracellular ATP levels in HNSCC cells, at least in part, through its inhibitory effect on the mitochondrial protein IF1. The addition of ATP attenuated CYT997-induced suppression of cell invasion, coupled with down-regulation of E-Cadherin and up-regulation of Vimentin. These findings support a critical role of ATP levels in cell invasion and metastasis under the influence of CYT997. Collectively, our data unveil the mechanism involved in mediating CYT997 action, and provide preclinical rationale for possible clinical application of CYT997 as a novel therapeutic strategy against aggressive HNSCC.
Collapse
Affiliation(s)
- Xiangdong Zhao
- Department of Otorhinolaryngology, Head and Neck SurgeryGuangdong Second Provincial General HospitalGuangzhouGuangdongChina
- Department of Oral Biology and Diagnostic SciencesDental College of GeorgiaAugusta UniversityAugustaGeorgia
| | - Liwei Lang
- Department of Oral Biology and Diagnostic SciencesDental College of GeorgiaAugusta UniversityAugustaGeorgia
| | - Leilei He
- Department of Oral Biology and Diagnostic SciencesDental College of GeorgiaAugusta UniversityAugustaGeorgia
| | - Lixia Gao
- Department of Oral Biology and Diagnostic SciencesDental College of GeorgiaAugusta UniversityAugustaGeorgia
| | - David Chyan
- Department of Oral Biology and Diagnostic SciencesDental College of GeorgiaAugusta UniversityAugustaGeorgia
- Department of BiologyCollege of Science and MathematicsAugusta UniversityAugustaGeorgia
| | - Yuanping Xiong
- Department of Oral Biology and Diagnostic SciencesDental College of GeorgiaAugusta UniversityAugustaGeorgia
| | - Honglin Li
- Department of Biochemistry and Molecular BiologyGeorgia Cancer CenterMedical College of GeorgiaAugusta UniversityAugustaGeorgia
| | - Hong Peng
- Department of Otorhinolaryngology, Head and Neck SurgeryGuangdong Second Provincial General HospitalGuangzhouGuangdongChina
| | - Yong Teng
- Department of Oral Biology and Diagnostic SciencesDental College of GeorgiaAugusta UniversityAugustaGeorgia
- Department of Biochemistry and Molecular BiologyGeorgia Cancer CenterMedical College of GeorgiaAugusta UniversityAugustaGeorgia
- Department of Medical Laboratory, Imaging and Radiologic SciencesCollege of Allied HealthAugusta UniversityAugustaGeorgia
| |
Collapse
|
28
|
Andjelković A, Mordas A, Bruinsma L, Ketola A, Cannino G, Giordano L, Dhandapani PK, Szibor M, Dufour E, Jacobs HT. Expression of the Alternative Oxidase Influences Jun N-Terminal Kinase Signaling and Cell Migration. Mol Cell Biol 2018; 38:e00110-18. [PMID: 30224521 PMCID: PMC6275184 DOI: 10.1128/mcb.00110-18] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 04/11/2018] [Accepted: 09/11/2018] [Indexed: 12/25/2022] Open
Abstract
Downregulation of Jun N-terminal kinase (JNK) signaling inhibits cell migration in diverse model systems. In Drosophila pupal development, attenuated JNK signaling in the thoracic dorsal epithelium leads to defective midline closure, resulting in cleft thorax. Here we report that concomitant expression of the Ciona intestinalis alternative oxidase (AOX) was able to compensate for JNK pathway downregulation, substantially correcting the cleft thorax phenotype. AOX expression also promoted wound-healing behavior and single-cell migration in immortalized mouse embryonic fibroblasts (iMEFs), counteracting the effect of JNK pathway inhibition. However, AOX was not able to rescue developmental phenotypes resulting from knockdown of the AP-1 transcription factor, the canonical target of JNK, nor its targets and had no effect on AP-1-dependent transcription. The migration of AOX-expressing iMEFs in the wound-healing assay was differentially stimulated by antimycin A, which redirects respiratory electron flow through AOX, altering the balance between mitochondrial ATP and heat production. Since other treatments affecting mitochondrial ATP did not stimulate wound healing, we propose increased mitochondrial heat production as the most likely primary mechanism of action of AOX in promoting cell migration in these various contexts.
Collapse
Affiliation(s)
- Ana Andjelković
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Amelia Mordas
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Lyon Bruinsma
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Annika Ketola
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Giuseppe Cannino
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Luca Giordano
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Praveen K Dhandapani
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Marten Szibor
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Eric Dufour
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Howard T Jacobs
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
29
|
Fitzgerald G, Soro-Arnaiz I, De Bock K. The Warburg Effect in Endothelial Cells and its Potential as an Anti-angiogenic Target in Cancer. Front Cell Dev Biol 2018; 6:100. [PMID: 30255018 PMCID: PMC6141712 DOI: 10.3389/fcell.2018.00100] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 08/09/2018] [Indexed: 12/29/2022] Open
Abstract
Endothelial cells (ECs) make up the lining of our blood vessels and they ensure optimal nutrient and oxygen delivery to the parenchymal tissue. In response to oxygen and/or nutrient deprivation, ECs become activated and sprout into hypo-vascularized tissues forming new vascular networks in a process termed angiogenesis. New sprouts are led by migratory tip cells and extended through the proliferation of trailing stalk cells. Activated ECs rewire their metabolism to cope with the increased energetic and biosynthetic demands associated with migration and proliferation. Moreover, metabolic signaling pathways interact and integrate with angiogenic signaling events. These metabolic adaptations play essential roles in determining EC fate and function, and are perturbed during pathological angiogenesis, as occurs in cancer. The angiogenic switch, or the growth of new blood vessels into an expanding tumor, increases tumor growth and malignancy. Limiting tumor angiogenesis has therefore long been a goal for anticancer therapy but the traditional growth factor targeted anti-angiogenic treatments have met with limited success. In recent years however, it has become increasingly recognized that focusing on altered tumor EC metabolism provides an attractive alternative anti-angiogenic strategy. In this review, we will describe the EC metabolic signature and how changes in EC metabolism affect EC fate during physiological sprouting, as well as in the cancer setting. Then, we will discuss the potential of targeting EC metabolism as a promising approach to develop new anti-cancer therapies.
Collapse
Affiliation(s)
- Gillian Fitzgerald
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology Zurich, Zurich, Switzerland
| | - Inés Soro-Arnaiz
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology Zurich, Zurich, Switzerland
| | - Katrien De Bock
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology Zurich, Zurich, Switzerland
| |
Collapse
|
30
|
Adenylate kinase potentiates the capsular polysaccharide by modulating Cps2D in Streptococcus pneumoniae D39. Exp Mol Med 2018; 50:1-14. [PMID: 30185778 PMCID: PMC6123713 DOI: 10.1038/s12276-018-0141-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 05/05/2018] [Accepted: 05/21/2018] [Indexed: 01/20/2023] Open
Abstract
Streptococcus pneumoniae is a polysaccharide-encapsulated bacterium. The capsule thickens during blood invasion compared with the thinner capsules observed in asymptomatic nasopharyngeal colonization. However, the underlying mechanism regulating differential CPS expression remains unclear. CPS synthesis requires energy that is supplied by ATP. Previously, we demonstrated a correlation between ATP levels and adenylate kinase in S. pneumoniae (SpAdK). A dose-dependent induction of SpAdK in serum was also reported. To meet medical needs, this study aimed to elucidate the role of SpAdK in the regulation of CPS production. CPS levels in S. pneumoniae type 2 (D39) increased proportionally with SpAdK levels, but they were not related to pneumococcal autolysis. Moreover, increased SpAdK levels resulted in increased total tyrosine kinase Cps2D levels and phosphorylated Cps2D, which is a regulator of CPS synthesis in the D39 strain. Our results also indicated that the SpAdK and Cps2D proteins interact in the presence of Mg-ATP. In addition, in silico analysis uncovered the mechanism behind this protein–protein interaction, suggesting that SpAdK binds with the Cps2D dimer. This established the importance of the ATP-binding domain of Cps2D. Taken together, the biophysical interaction between SpAdK and Cps2D plays an important role in enhancing Cps2D phosphorylation, which results in increased CPS synthesis. A physical interaction between two key enzymes explains how the bacterium responsible for causing pneumococcal disease thickens its external capsule during infection of the bloodstream. A team led by Dong-Kwon Rhee from Sungkyunkwan University in Suwon, South Korea, studied strains of Streptococcus pneumoniae expressing varying levels of an enzyme that helps maintain the proper balance of cellular energy. They found that this enzyme stimulated the production of sugar chains that coat the outside of the bacterial capsule by binding and activating an intermediary enzyme involved in the synthesis of these sugar chains. Since the capsule is critical in warding off the human immune response, the findings suggest that drugs designed to disrupt the enzyme-mediated induction of capsule formation could help prevent or treat pneumococcal disease.
Collapse
|
31
|
Xu S, Catapang A, Braas D, Stiles L, Doh HM, Lee JT, Graeber TG, Damoiseaux R, Shirihai O, Herschman HR. A precision therapeutic strategy for hexokinase 1-null, hexokinase 2-positive cancers. Cancer Metab 2018; 6:7. [PMID: 29988332 PMCID: PMC6022704 DOI: 10.1186/s40170-018-0181-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 05/17/2018] [Indexed: 01/19/2023] Open
Abstract
Background Precision medicine therapies require identification of unique molecular cancer characteristics. Hexokinase (HK) activity has been proposed as a therapeutic target; however, different hexokinase isoforms have not been well characterized as alternative targets. While HK2 is highly expressed in the majority of cancers, cancer subtypes with differential HK1 and HK2 expression have not been characterized for their sensitivities to HK2 silencing. Methods HK1 and HK2 expression in the Cancer Cell Line Encyclopedia dataset was analyzed. A doxycycline-inducible shRNA silencing system was used to examine the effect of HK2 knockdown in cultured cells and in xenograft models of HK1−HK2+ and HK1+HK2+ cancers. Glucose consumption and lactate production rates were measured to monitor HK activity in cell culture, and 18F-FDG PET/CT was used to monitor HK activity in xenograft tumors. A high-throughput screen was performed to search for synthetically lethal compounds in combination with HK2 inhibition in HK1−HK2+ liver cancer cells, and a combination therapy for liver cancers with this phenotype was developed. A metabolomic analysis was performed to examine changes in cellular energy levels and key metabolites in HK1−HK2+ cells treated with this combination therapy. The CRISPR Cas9 method was used to establish isogenic HK1+HK2+ and HK1−HK2+ cell lines to evaluate HK1−HK2+ cancer cell sensitivity to the combination therapy. Results Most tumors express both HK1 and HK2, and subsets of cancers from a wide variety of tissues of origin express only HK2. Unlike HK1+HK2+ cancers, HK1−HK2+ cancers are sensitive to HK2 silencing-induced cytostasis. Synthetic lethality was achieved in HK1−HK2+ liver cancer cells, by the combination of DPI, a mitochondrial complex I inhibitor, and HK2 inhibition, in HK1−HK2+ liver cancer cells. Perhexiline, a fatty acid oxidation inhibitor, further sensitizes HK1−HK2+ liver cancer cells to the complex I/HK2-targeted therapeutic combination. Although HK1+HK2+ lung cancer H460 cells are resistant to this therapeutic combination, isogenic HK1KOHK2+ cells are sensitive to this therapy. Conclusions The HK1−HK2+ cancer subsets exist among a wide variety of cancer types. Selective inhibition of the HK1−HK2+ cancer cell-specific energy production pathways (HK2-driven glycolysis, oxidative phosphorylation and fatty acid oxidation), due to the unique presence of only the HK2 isoform, appears promising to treat HK1−HK2+ cancers. This therapeutic strategy will likely be tolerated by most normal tissues, where only HK1 is expressed. Electronic supplementary material The online version of this article (10.1186/s40170-018-0181-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shili Xu
- 1Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 USA
| | - Arthur Catapang
- 1Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 USA
| | - Daniel Braas
- 1Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 USA.,3UCLA Metabolomics Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 USA
| | - Linsey Stiles
- 6Division of Endocrinology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 USA
| | - Hanna M Doh
- 1Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 USA
| | - Jason T Lee
- 1Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 USA.,4Crump Institute for Molecular Imaging, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 USA.,5Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 USA
| | - Thomas G Graeber
- 1Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 USA.,3UCLA Metabolomics Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 USA.,4Crump Institute for Molecular Imaging, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 USA.,5Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 USA
| | - Robert Damoiseaux
- 1Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 USA.,7California NanoSystems Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 USA
| | - Orian Shirihai
- 6Division of Endocrinology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 USA
| | - Harvey R Herschman
- 1Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 USA.,2Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 USA.,4Crump Institute for Molecular Imaging, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 USA.,5Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 USA.,8Molecular Biology Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 USA
| |
Collapse
|
32
|
de la Ballina NR, Villalba A, Cao A. Proteomic profile of Ostrea edulis haemolymph in response to bonamiosis and identification of candidate proteins as resistance markers. DISEASES OF AQUATIC ORGANISMS 2018; 128:127-145. [PMID: 29733027 DOI: 10.3354/dao03220] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
European flat oyster Ostrea edulis populations have suffered extensive mortalities caused by bonamiosis. The protozoan parasite Bonamia ostreae is largely responsible for this disease in Europe, while its congener B. exitiosa has been detected more recently in various European countries. Both of these intracellular parasites are able to survive and proliferate within haemocytes, the main cellular effectors of the immune system in molluscs. Two-dimensional electrophoresis was used to compare the haemolymph protein profile between Bonamia spp.-infected and non-infected oysters within 3 different stocks, a Galician stock of oysters selected for resistance against bonamiosis, a non-selected Galician stock and a selected Irish stock. Thirty-four proteins with a presumably relevant role in the oyster-Bonamia spp. interaction were identified; they were involved in major metabolic pathways, such as energy production, respiratory chain, oxidative stress, signal transduction, transcription, translation, protein degradation and cell defence. Furthermore, the haemolymph proteomic profiles of the non-infected oysters of the 2 Galician stocks were compared. As a result, 7 proteins representative of the non-infected Galician oysters selected for resistance against bonamiosis were identified; these 7 proteins could be considered as candidate markers of resistance to bonamiosis, which should be further assessed.
Collapse
Affiliation(s)
- Nuria R de la Ballina
- Centro de Investigacións Mariñas (CIMA), Consellería do Mar, Xunta de Galicia, 36620 Vilanova de Arousa, Spain
| | | | | |
Collapse
|
33
|
Zanotelli MR, Goldblatt ZE, Miller JP, Bordeleau F, Li J, VanderBurgh JA, Lampi MC, King MR, Reinhart-King CA. Regulation of ATP utilization during metastatic cell migration by collagen architecture. Mol Biol Cell 2018; 29:1-9. [PMID: 29118073 PMCID: PMC5746062 DOI: 10.1091/mbc.e17-01-0041] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 10/17/2017] [Accepted: 11/03/2017] [Indexed: 01/04/2023] Open
Abstract
Cell migration in a three-dimensional matrix requires that cells either remodel the surrounding matrix fibers and/or squeeze between the fibers to move. Matrix degradation, matrix remodeling, and changes in cell shape each require cells to expend energy. While significant research has been performed to understand the cellular and molecular mechanisms guiding metastatic migration, less is known about cellular energy regulation and utilization during three-dimensional cancer cell migration. Here we introduce the use of the genetically encoded fluorescent biomarkers, PercevalHR and pHRed, to quantitatively assess ATP, ADP, and pH levels in MDA-MB-231 metastatic cancer cells as a function of the local collagen microenvironment. We find that the use of the probe is an effective tool for exploring the thermodynamics of cancer cell migration and invasion. Specifically, we find that the ATP:ADP ratio increases in cells in denser matrices, where migration is impaired, and it decreases in cells in aligned collagen matrices, where migration is facilitated. When migration is pharmacologically inhibited, the ATP:ADP ratio decreases. Together, our data indicate that matrix architecture alters cellular energetics and that intracellular ATP:ADP ratio is related to the ability of cancer cells to effectively migrate.
Collapse
Affiliation(s)
- Matthew R Zanotelli
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853
| | - Zachary E Goldblatt
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853
| | - Joseph P Miller
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853
| | - Francois Bordeleau
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37212
| | - Jiahe Li
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853
| | - Jacob A VanderBurgh
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853
| | - Marsha C Lampi
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853
| | - Michael R King
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37212
| | - Cynthia A Reinhart-King
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37212
| |
Collapse
|
34
|
Colquhoun A. Cell biology-metabolic crosstalk in glioma. Int J Biochem Cell Biol 2017; 89:171-181. [PMID: 28549626 DOI: 10.1016/j.biocel.2017.05.022] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 05/15/2017] [Accepted: 05/17/2017] [Indexed: 12/19/2022]
Abstract
The renewed interest in cancer metabolism in recent years has been fuelled by the identification of the involvement of key oncogenes and tumour suppressor genes in the control of metabolic pathways. Many of these alterations lead to dramatic changes in bioenergetics, biosynthesis and redox balance within tumour cells. The complex relationship between tumour cell metabolism and the tumour microenvironment has turned this field of biochemistry and cell biology into a challenging and exciting area for study. In the case of gliomas the involvement of altered metabolic pathways including glycolysis, oxidative phosphorylation and glutaminolysis are pointing the way to new possibilities for treatment. The tumour-promoting effects of inflammation are an emerging hallmark of cancer and the role of the eicosanoids in gliomas is an area of active research to elucidate the importance of individual eicosanoids in glioma cell proliferation, migration and immune escape. In this review, the different aspects of metabolic reprogramming which occur in gliomas are highlighted and their relationship to glioma cell biology and the wider tumour microenvironment is described.
Collapse
Affiliation(s)
- Alison Colquhoun
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
35
|
Zala D, Schlattner U, Desvignes T, Bobe J, Roux A, Chavrier P, Boissan M. The advantage of channeling nucleotides for very processive functions. F1000Res 2017; 6:724. [PMID: 28663786 PMCID: PMC5473427 DOI: 10.12688/f1000research.11561.2] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/17/2017] [Indexed: 12/26/2022] Open
Abstract
Nucleoside triphosphate (NTP)s, like ATP (adenosine 5'-triphosphate) and GTP (guanosine 5'-triphosphate), have long been considered sufficiently concentrated and diffusible to fuel all cellular ATPases (adenosine triphosphatases) and GTPases (guanosine triphosphatases) in an energetically healthy cell without becoming limiting for function. However, increasing evidence for the importance of local ATP and GTP pools, synthesised in close proximity to ATP- or GTP-consuming reactions, has fundamentally challenged our view of energy metabolism. It has become evident that cellular energy metabolism occurs in many specialised 'microcompartments', where energy in the form of NTPs is transferred preferentially from NTP-generating modules directly to NTP-consuming modules. Such energy channeling occurs when diffusion through the cytosol is limited, where these modules are physically close and, in particular, if the NTP-consuming reaction has a very high turnover, i.e. is very processive. Here, we summarise the evidence for these conclusions and describe new insights into the physiological importance and molecular mechanisms of energy channeling gained from recent studies. In particular, we describe the role of glycolytic enzymes for axonal vesicle transport and nucleoside diphosphate kinases for the functions of dynamins and dynamin-related GTPases.
Collapse
Affiliation(s)
- Diana Zala
- ESPCI - Paris, PSL Research University, Paris, F-75005, France.,CNRS, UMR8249, Paris, F-75005, France
| | - Uwe Schlattner
- Laboratory of Fundamental and Applied Bioenergetics (LBFA), and SFR Environmental and Systems Biology (BEeSy), U1055, University Grenoble Alpes, Grenoble, 38058, France.,Inserm-U1055, Grenoble, F-38058, France
| | - Thomas Desvignes
- Institute of Neuroscience, University of Oregon, Eugene, OR, 97401, USA
| | - Julien Bobe
- INRA, UR1037 LPGP, Campus de Beaulieu, Rennes, F-35000, France
| | - Aurélien Roux
- Department of Biochemistry, University of Geneva, Geneva, CH-1211, Switzerland.,Swiss National Centre for Competence in Research Programme Chemical Biology, Geneva, CH-1211, Switzerland
| | - Philippe Chavrier
- Institut Curie, Paris, F-75248, France.,PSL Research University, Paris, F-75005, France.,CNRS, UMR144, Paris, F-75248, France
| | - Mathieu Boissan
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, UMRS938, Saint-Antoine Research Center, Paris, F-75012, France.,AP-HP, Hospital Tenon, Service de Biochimie et Hormonologie, Paris, F-75020, France
| |
Collapse
|
36
|
Zala D, Schlattner U, Desvignes T, Bobe J, Roux A, Chavrier P, Boissan M. The advantage of channeling nucleotides for very processive functions. F1000Res 2017; 6:724. [PMID: 28663786 DOI: 10.12688/f1000research.11561.1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/15/2017] [Indexed: 01/01/2023] Open
Abstract
Nucleoside triphosphate (NTP)s, like ATP (adenosine 5'-triphosphate) and GTP (guanosine 5'-triphosphate), have long been considered sufficiently concentrated and diffusible to fuel all cellular ATPases (adenosine triphosphatases) and GTPases (guanosine triphosphatases) in an energetically healthy cell without becoming limiting for function. However, increasing evidence for the importance of local ATP and GTP pools, synthesised in close proximity to ATP- or GTP-consuming reactions, has fundamentally challenged our view of energy metabolism. It has become evident that cellular energy metabolism occurs in many specialised 'microcompartments', where energy in the form of NTPs is transferred preferentially from NTP-generating modules directly to NTP-consuming modules. Such energy channeling occurs when diffusion through the cytosol is limited, where these modules are physically close and, in particular, if the NTP-consuming reaction has a very high turnover, i.e. is very processive. Here, we summarise the evidence for these conclusions and describe new insights into the physiological importance and molecular mechanisms of energy channeling gained from recent studies. In particular, we describe the role of glycolytic enzymes for axonal vesicle transport and nucleoside diphosphate kinases for the functions of dynamins and dynamin-related GTPases.
Collapse
Affiliation(s)
- Diana Zala
- ESPCI - Paris, PSL Research University, Paris, F-75005, France.,CNRS, UMR8249, Paris, F-75005, France
| | - Uwe Schlattner
- Laboratory of Fundamental and Applied Bioenergetics (LBFA), and SFR Environmental and Systems Biology (BEeSy), U1055, University Grenoble Alpes, Grenoble, 38058, France.,Inserm-U1055, Grenoble, F-38058, France
| | - Thomas Desvignes
- Institute of Neuroscience, University of Oregon, Eugene, OR, 97401, USA
| | - Julien Bobe
- INRA, UR1037 LPGP, Campus de Beaulieu, Rennes, F-35000, France
| | - Aurélien Roux
- Department of Biochemistry, University of Geneva, Geneva, CH-1211, Switzerland.,Swiss National Centre for Competence in Research Programme Chemical Biology, Geneva, CH-1211, Switzerland
| | - Philippe Chavrier
- Institut Curie, Paris, F-75248, France.,PSL Research University, Paris, F-75005, France.,CNRS, UMR144, Paris, F-75248, France
| | - Mathieu Boissan
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, UMRS938, Saint-Antoine Research Center, Paris, F-75012, France.,AP-HP, Hospital Tenon, Service de Biochimie et Hormonologie, Paris, F-75020, France
| |
Collapse
|
37
|
Microphthalmia-associated transcription factor suppresses invasion by reducing intracellular GTP pools. Oncogene 2016; 36:84-96. [PMID: 27181209 PMCID: PMC5112150 DOI: 10.1038/onc.2016.178] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 03/18/2016] [Accepted: 04/03/2016] [Indexed: 12/14/2022]
Abstract
Melanoma progression is associated with increased invasion and, often, decreased levels of microphthalmia-associated transcription factor (MITF). Accordingly, downregulation of MITF induces invasion in melanoma cells, however little is known about the underlying mechanisms. Here, we report for the first time that depletion of MITF results in elevation of intracellular GTP levels and increased amounts of active (GTP-bound) RAC1, RHO-A and RHO-C. Concomitantly, MITF-depleted cells display larger number of invadopodia and increased invasion. We further demonstrate that the gene for guanosine monophosphate reductase (GMPR) is a direct MITF target, and that the partial repression of GMPR accounts mostly for the above phenotypes in MITF-depleted cells. Reciprocally, transactivation of GMPR is required for MITF-dependent suppression of melanoma cell invasion, tumorigenicity, and lung colonization. Moreover, loss of GMPR accompanies downregulation of MITF in vemurafenib-resistant BRAFV600E-melanoma cells and underlies the increased invasion in these cells. Our data uncover novel mechanisms linking MITF-dependent inhibition of invasion to suppression of guanylate metabolism.
Collapse
|
38
|
Abstract
During mitosis, cells undergo massive deformation and reorganization, impacting on all cellular structures. Mitochondria, in particular, are highly dynamic organelles, which constantly undergo events of fission, fusion and cytoskeleton-based transport. This plasticity ensures the proper distribution of the metabolism, and the proper inheritance of functional organelles. During cell cycle, mitochondria undergo dramatic changes in distribution. In this review, we focus on the dynamic events that target mitochondria during mitosis. We describe how the cell-cycle-dependent microtubule-associated protein centromeric protein F (Cenp-F) is recruited to mitochondria by the mitochondrial Rho GTPase (Miro) to promote mitochondrial transport and re-distribution following cell division.
Collapse
|
39
|
Bianchi-Smiraglia A, Nikiforov MA. Slowing down the Grand Touring Prototype speed of cancer cells. Mol Cell Oncol 2016; 3. [PMID: 26989767 DOI: 10.1080/23723556.2015.1052181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Enzymes involved in de novo production of guanosine triphosphate (GTP) have been recently revealed as integral components of melanoma progression through modulation of the activity of small GTPases. Here, we discuss the biology and therapeutic implications of these findings.
Collapse
Affiliation(s)
- Anna Bianchi-Smiraglia
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York, 14263
| | - Mikhail A Nikiforov
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York, 14263
| |
Collapse
|
40
|
Nemutlu E, Gupta A, Zhang S, Viqar M, Holmuhamedov E, Terzic A, Jahangir A, Dzeja P. Decline of Phosphotransfer and Substrate Supply Metabolic Circuits Hinders ATP Cycling in Aging Myocardium. PLoS One 2015; 10:e0136556. [PMID: 26378442 PMCID: PMC4574965 DOI: 10.1371/journal.pone.0136556] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 08/04/2015] [Indexed: 12/24/2022] Open
Abstract
Integration of mitochondria with cytosolic ATP-consuming/ATP-sensing and substrate supply processes is critical for muscle bioenergetics and electrical activity. Whether age-dependent muscle weakness and increased electrical instability depends on perturbations in cellular energetic circuits is unknown. To define energetic remodeling of aged atrial myocardium we tracked dynamics of ATP synthesis-utilization, substrate supply, and phosphotransfer circuits through adenylate kinase (AK), creatine kinase (CK), and glycolytic/glycogenolytic pathways using 18O stable isotope-based phosphometabolomic technology. Samples of intact atrial myocardium from adult and aged rats were subjected to 18O-labeling procedure at resting basal state, and analyzed using the 18O-assisted HPLC-GC/MS technique. Characteristics for aging atria were lower inorganic phosphate Pi[18O], γ-ATP[18O], β-ADP[18O], and creatine phosphate CrP[18O] 18O-labeling rates indicating diminished ATP utilization-synthesis and AK and CK phosphotransfer fluxes. Shift in dynamics of glycolytic phosphotransfer was reflected in the diminished G6P[18O] turnover with relatively constant glycogenolytic flux or G1P[18O] 18O-labeling. Labeling of G3P[18O], an indicator of G3P-shuttle activity and substrate supply to mitochondria, was depressed in aged myocardium. Aged atrial myocardium displayed reduced incorporation of 18O into second (18O2), third (18O3), and fourth (18O4) positions of Pi[18O] and a lower Pi[18O]/γ-ATP[18 O]-labeling ratio, indicating delayed energetic communication and ATP cycling between mitochondria and cellular ATPases. Adrenergic stress alleviated diminished CK flux, AK catalyzed β-ATP turnover and energetic communication in aging atria. Thus, 18O-assisted phosphometabolomics uncovered simultaneous phosphotransfer through AK, CK, and glycolytic pathways and G3P substrate shuttle deficits hindering energetic communication and ATP cycling, which may underlie energetic vulnerability of aging atrial myocardium.
Collapse
Affiliation(s)
- Emirhan Nemutlu
- Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Anu Gupta
- Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Song Zhang
- Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Maria Viqar
- Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Ekhson Holmuhamedov
- Center for Integrative Research on Cardiovascular Aging (CIRCA), Aurora University of Wisconsin Medical Group, Aurora Health Care, Milwaukee, Wisconsin, United States of America
| | - Andre Terzic
- Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Arshad Jahangir
- Center for Integrative Research on Cardiovascular Aging (CIRCA), Aurora University of Wisconsin Medical Group, Aurora Health Care, Milwaukee, Wisconsin, United States of America
- * E-mail: (PD); (AJ)
| | - Petras Dzeja
- Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail: (PD); (AJ)
| |
Collapse
|
41
|
Das AM, Eggermont AMM, ten Hagen TLM. A ring barrier–based migration assay to assess cell migration in vitro. Nat Protoc 2015; 10:904-15. [DOI: 10.1038/nprot.2015.056] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
42
|
Metabolic pathway compartmentalization: an underappreciated opportunity? Curr Opin Biotechnol 2014; 34:73-81. [PMID: 25499800 DOI: 10.1016/j.copbio.2014.11.022] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 11/23/2014] [Indexed: 12/12/2022]
Abstract
For eukaryotic cells to function properly, they divide their intracellular space in subcellular compartments, each harboring specific metabolic activities. In recent years, it has become increasingly clear that compartmentalization of metabolic pathways is a prerequisite for certain cellular functions. This has for instance been documented for cellular migration, which relies on subcellular localization of glycolysis or mitochondrial respiration in a cell type-dependent manner. Although exciting, this field is still in its infancy, partly due to the limited availability of methods to study the directionality of metabolic pathways and to visualize metabolic processes in distinct cellular compartments. Nonetheless, advances in this field may offer opportunities for innovative strategies to target deregulated compartmentalized metabolism in disease.
Collapse
|
43
|
Thach TT, Lee S. New crystal structures of adenylate kinase from Streptococcus pneumoniae D39 in two conformations. Acta Crystallogr F Struct Biol Commun 2014; 70:1468-71. [PMID: 25372811 PMCID: PMC4231846 DOI: 10.1107/s2053230x14020718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 09/15/2014] [Indexed: 11/10/2022] Open
Abstract
Adenylate kinases (AdKs; EC 2.7.3.4) play a critical role in intercellular homeostasis by the interconversion of ATP and AMP to two ADP molecules. Crystal structures of adenylate kinase from Streptococcus pneumoniae D39 (SpAdK) have recently been determined using ligand-free and inhibitor-bound crystals belonging to space groups P21 and P1, respectively. Here, new crystal structures of SpAdK in ligand-free and inhibitor-bound states determined at 1.96 and 1.65 Å resolution, respectively, are reported. The new ligand-free crystal belonged to space group C2, with unit-cell parameters a=73.5, b=54.3, c=62.7 Å, β=118.8°. The new ligand-free structure revealed an open conformation that differed from the previously determined conformation, with an r.m.s.d on Cα atoms of 1.4 Å. The new crystal of the complex with the two-substrate-mimicking inhibitor P1,P5-bis(adenosine-5'-)pentaphosphate (Ap5A) belonged to space group P1, with unit-cell parameters a=53.9, b=62.3, c=63.0 Å, α=101.9, β=112.6, γ=89.9°. Despite belonging to the same space group as the previously reported crystal, the new Ap5A-bound crystal contains four molecules in the asymmetric unit, compared with two in the previous crystal, and shows slightly different lattice contacts. These results demonstrate that SpAdK can crystallize promiscuously in different forms and that the open structure is flexible in conformation.
Collapse
Affiliation(s)
- Trung Thanh Thach
- Department of Biological Sciences, Sungkyunkwan University, 2066 Seobu-ro, Suwon, Gyeonggi 440-746, Republic of Korea
| | - Sangho Lee
- Department of Biological Sciences, Sungkyunkwan University, 2066 Seobu-ro, Suwon, Gyeonggi 440-746, Republic of Korea
| |
Collapse
|
44
|
Venter G, Oerlemans FTJJ, Willemse M, Wijers M, Fransen JAM, Wieringa B. NAMPT-mediated salvage synthesis of NAD+ controls morphofunctional changes of macrophages. PLoS One 2014; 9:e97378. [PMID: 24824795 PMCID: PMC4019579 DOI: 10.1371/journal.pone.0097378] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 04/18/2014] [Indexed: 12/31/2022] Open
Abstract
Functional morphodynamic behavior of differentiated macrophages is strongly controlled by actin cytoskeleton rearrangements, a process in which also metabolic cofactors ATP and NAD(H) (i.e. NAD+ and NADH) and NADP(H) (i.e. NADP+ and NADPH) play an essential role. Whereas the link to intracellular ATP availability has been studied extensively, much less is known about the relationship between actin cytoskeleton dynamics and intracellular redox state and NAD+-supply. Here, we focus on the role of nicotinamide phosphoribosyltransferase (NAMPT), found in extracellular form as a cytokine and growth factor, and in intracellular form as one of the key enzymes for the production of NAD+ in macrophages. Inhibition of NAD+ salvage synthesis by the NAMPT-specific drug FK866 caused a decrease in cytosolic NAD+ levels in RAW 264.7 and Maf-DKO macrophages and led to significant downregulation of the glycolytic flux without directly affecting cell viability, proliferation, ATP production capacity or mitochondrial respiratory activity. Concomitant with these differential metabolic changes, the capacity for phagocytic ingestion of particles and also substrate adhesion of macrophages were altered. Depletion of cytoplasmic NAD+ induced cell-morphological changes and impaired early adhesion in phagocytosis of zymosan particles as well as spreading performance. Restoration of NAD+ levels by NAD+, NMN, or NADP+ supplementation reversed the inhibitory effects of FK866. We conclude that direct coupling to local, actin-based, cytoskeletal dynamics is an important aspect of NAD+'s cytosolic role in the regulation of morphofunctional characteristics of macrophages.
Collapse
Affiliation(s)
- Gerda Venter
- Department of Cell Biology, Nijmegen Centre for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Frank T. J. J. Oerlemans
- Department of Cell Biology, Nijmegen Centre for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Marieke Willemse
- Department of Cell Biology, Nijmegen Centre for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Mietske Wijers
- Department of Cell Biology, Nijmegen Centre for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Jack A. M. Fransen
- Department of Cell Biology, Nijmegen Centre for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Bé Wieringa
- Department of Cell Biology, Nijmegen Centre for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
45
|
Venter G, Oerlemans FTJJ, Wijers M, Willemse M, Fransen JAM, Wieringa B. Glucose controls morphodynamics of LPS-stimulated macrophages. PLoS One 2014; 9:e96786. [PMID: 24796786 PMCID: PMC4010488 DOI: 10.1371/journal.pone.0096786] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 04/11/2014] [Indexed: 12/12/2022] Open
Abstract
Macrophages constantly undergo morphological changes when quiescently surveying the tissue milieu for signs of microbial infection or damage, or after activation when they are phagocytosing cellular debris or foreign material. These morphofunctional alterations require active actin cytoskeleton remodeling and metabolic adaptation. Here we analyzed RAW 264.7 and Maf-DKO macrophages as models to study whether there is a specific association between aspects of carbohydrate metabolism and actin-based processes in LPS-stimulated macrophages. We demonstrate that the capacity to undergo LPS-induced cell shape changes and to phagocytose complement-opsonized zymosan (COZ) particles does not depend on oxidative phosphorylation activity but is fueled by glycolysis. Different macrophage activities like spreading, formation of cell protrusions, as well as phagocytosis of COZ, were thereby strongly reliant on the presence of low levels of extracellular glucose. Since global ATP production was not affected by rewiring of glucose catabolism and inhibition of glycolysis by 2-deoxy-D-glucose and glucose deprivation had differential effects, our observations suggest a non-metabolic role for glucose in actin cytoskeletal remodeling in macrophages, e.g. via posttranslational modification of receptors or signaling molecules, or other effects on the machinery that drives actin cytoskeletal changes. Our findings impute a decisive role for the nutrient state of the tissue microenvironment in macrophage morphodynamics.
Collapse
Affiliation(s)
- Gerda Venter
- Department of Cell Biology, Nijmegen Centre for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Frank T. J. J. Oerlemans
- Department of Cell Biology, Nijmegen Centre for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Mietske Wijers
- Department of Cell Biology, Nijmegen Centre for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Marieke Willemse
- Department of Cell Biology, Nijmegen Centre for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Jack A. M. Fransen
- Department of Cell Biology, Nijmegen Centre for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Bé Wieringa
- Department of Cell Biology, Nijmegen Centre for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
46
|
Adenylate Kinase Isoform Network: A Major Hub in Cell Energetics and Metabolic Signaling. SYSTEMS BIOLOGY OF METABOLIC AND SIGNALING NETWORKS 2014. [DOI: 10.1007/978-3-642-38505-6_6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
47
|
Wawrzyniak JA, Bianchi-Smiraglia A, Bshara W, Mannava S, Ackroyd J, Bagati A, Omilian AR, Im M, Fedtsova N, Miecznikowski JC, Moparthy KC, Zucker SN, Zhu Q, Kozlova NI, Berman AE, Hoek KS, Gudkov AV, Shewach DS, Morrison CD, Nikiforov MA. A purine nucleotide biosynthesis enzyme guanosine monophosphate reductase is a suppressor of melanoma invasion. Cell Rep 2013; 5:493-507. [PMID: 24139804 DOI: 10.1016/j.celrep.2013.09.015] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 08/20/2013] [Accepted: 09/11/2013] [Indexed: 01/02/2023] Open
Abstract
Melanoma is one of the most aggressive types of human cancers, and the mechanisms underlying melanoma invasive phenotype are not completely understood. Here, we report that expression of guanosine monophosphate reductase (GMPR), an enzyme involved in de novo biosynthesis of purine nucleotides, was downregulated in the invasive stages of human melanoma. Loss- and gain-of-function experiments revealed that GMPR downregulates the amounts of several GTP-bound (active) Rho-GTPases and suppresses the ability of melanoma cells to form invadopodia, degrade extracellular matrix, invade in vitro, and grow as tumor xenografts in vivo. Mechanistically, we demonstrated that GMPR partially depletes intracellular GTP pools. Pharmacological inhibition of de novo GTP biosynthesis suppressed whereas addition of exogenous guanosine increased invasion of melanoma cells as well as cells from other cancer types. Our data identify GMPR as a melanoma invasion suppressor and establish a link between guanosine metabolism and Rho-GTPase-dependent melanoma cell invasion.
Collapse
Affiliation(s)
- Joseph A Wawrzyniak
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
van Horssen R, Willemse M, Haeger A, Attanasio F, Güneri T, Schwab A, Stock CM, Buccione R, Fransen JAM, Wieringa B. Intracellular NAD(H) levels control motility and invasion of glioma cells. Cell Mol Life Sci 2013; 70:2175-90. [PMID: 23307072 PMCID: PMC11113314 DOI: 10.1007/s00018-012-1249-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Revised: 11/29/2012] [Accepted: 12/17/2012] [Indexed: 12/15/2022]
Abstract
Oncogenic transformation involves reprogramming of cell metabolism, whereby steady-state levels of intracellular NAD(+) and NADH can undergo dramatic changes while ATP concentration is generally well maintained. Altered expression of nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme of NAD(+)-salvage, accompanies the changes in NAD(H) during tumorigenesis. Here, we show by genetic and pharmacological inhibition of NAMPT in glioma cells that fluctuation in intracellular [NAD(H)] differentially affects cell growth and morphodynamics, with motility/invasion capacity showing the highest sensitivity to [NAD(H)] decrease. Extracellular supplementation of NAD(+) or re-expression of NAMPT abolished the effects. The effects of NAD(H) decrease on cell motility appeared parallel coupled with diminished pyruvate-lactate conversion by lactate dehydrogenase (LDH) and with changes in intracellular and extracellular pH. The addition of lactic acid rescued and knockdown of LDH-A replicated the effects of [NAD(H)] on motility. Combined, our observations demonstrate that [NAD(H)] is an important metabolic component of cancer cell motility. Nutrient or drug-mediated modulation of NAD(H) levels may therefore represent a new option for blocking the invasive behavior of tumors.
Collapse
Affiliation(s)
- Remco van Horssen
- Department of Cell Biology, Nijmegen Centre for Molecular Life Sciences (NCMLS), Radboud University Nijmegen Medical Centre, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
van Horssen R, Buccione R, Willemse M, Cingir S, Wieringa B, Attanasio F. Cancer cell metabolism regulates extracellular matrix degradation by invadopodia. Eur J Cell Biol 2013; 92:113-21. [PMID: 23306026 DOI: 10.1016/j.ejcb.2012.11.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Revised: 11/20/2012] [Accepted: 11/27/2012] [Indexed: 11/19/2022] Open
Abstract
Transformed cancer cells have an altered metabolism, characterized by a shift towards aerobic glycolysis, referred to as 'the Warburg phenotype'. A change in flux through mitochondrial OXPHOS and cytosolic pathways for ATP production and a gain of capacity for biomass production in order to sustain the needs for altered growth and morphodynamics are typically involved in this global rewiring of cancer cell metabolism. Characteristically, these changes in metabolism are accompanied by enhanced uptake of nutrients like glucose and glutamine. Here we focus on the relationship between cell metabolism and cell dynamics, in particular the formation and function of invadopodia, specialized structures for focal degradation of the extracellular matrix. Since we recently found presence of enzymes that are active in glycolysis and associated pathways in invadopodia, we hypothesize that metabolic adaptation and invadopodia formation are linked processes. We give an overview on the background for this idea and show for the first time that extracellular matrix degradation by invadopodia can be differentially manipulated, without effects on cell proliferation, by use of metabolic inhibitors or changes in nutrient composition of cell culture media. We conclude that cell metabolism and carbohydrate availability, especially pyruvate, are involved in fuelling of invadopodia formation and activity.
Collapse
Affiliation(s)
- Remco van Horssen
- Department of Cell Biology, Nijmegen Centre for Molecular Life Sciences (NCMLS), Radboud University Medical Centre, Nijmegen, The Netherlands.
| | | | | | | | | | | |
Collapse
|
50
|
Abstract
Although cancer is a diverse set of diseases, cancer cells share a number of adaptive hallmarks. Dysregulated pH is emerging as a hallmark of cancer because cancers show a 'reversed' pH gradient with a constitutively increased intracellular pH that is higher than the extracellular pH. This gradient enables cancer progression by promoting proliferation, the evasion of apoptosis, metabolic adaptation, migration and invasion. Several new advances, including an increased understanding of pH sensors, have provided insight into the molecular basis for pH-dependent cell behaviours that are relevant to cancer cell biology. We highlight the central role of pH sensors in cancer cell adaptations and suggest how dysregulated pH could be exploited to develop cancer-specific therapeutics.
Collapse
Affiliation(s)
- Bradley A Webb
- Department of Cell and Tissue Biology, University of California, San Francisco, California 94143, USA
| | | | | | | |
Collapse
|