1
|
Samareh B, Klimenkova O, Aghaallaei N, Cheng L, Zikic A, Loghmani H, Tesakov I, Müller P, Suttorp M, Welte K, Skokowa J, Morishima T. NAMPT-mediated deacetylation of HCLS1 protein promotes clonogenic growth of pediatric CML cells. Exp Hematol 2025:104801. [PMID: 40349748 DOI: 10.1016/j.exphem.2025.104801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 04/21/2025] [Accepted: 04/30/2025] [Indexed: 05/14/2025]
Abstract
Pediatric chronic myeloid leukemia (CML) is a rare hematologic malignancy with biological features that differ from that of adult patients. In pediatric patients with CML the burden of tumor cells is higher resulting in a delayed achievement of deep molecular response (DMR) upon treatment with tyrosine kinase inhibitors (TKIs, e.g., imatinib) than what has been reported in adults. Therefore, the probability to develop resistance to TKIs in children with CML is higher than in adults due to much longer exposure to TKIs. Moreover, in children with CML, long-term treatment with imatinib causes hematologic and nonhematologic toxicities. Improvements of CML therapy in pediatric patients based on the targeting of hematopoiesis-specific BCR::ABL1 downstream effectors are needed. Here, we report elevated levels of the nicotinamide phosphoribosyltransferase (NAMPT) in mononuclear cells of pediatric patients with chronic phase CML (CP-CML) and in blastic phase CML cell lines. NAMPT inhibition abrogated in vitro clonogenic capacity and proliferation of CML cells. NAMPT deacetylates and activates the hematopoietic-specific lyn-substrate 1 (HCLS1) protein, which is essential for the proliferation of CML cells. Moreover, IL1RAP - a marker of myeloid leukemia-initiating cells - and LEF-1 - a transcription factor of Wnt signaling - are downstream targets of NAMPT/HCLS1 pathway. Together, our results reveal new treatment avenues of pediatric patients with CML by targeting NAMPT-mediated deacetylation of the hematopoietic-specific HCLS1 protein.
Collapse
Affiliation(s)
- Bardia Samareh
- Department of Hematology, Oncology, Immunology, Rheumatology and Pulmonology, University Hospital Tübingen, Tübingen, Germany
| | - Olga Klimenkova
- Department of Hematology, Oncology, Immunology, Rheumatology and Pulmonology, University Hospital Tübingen, Tübingen, Germany
| | - Narges Aghaallaei
- Department of Hematology, Oncology, Immunology, Rheumatology and Pulmonology, University Hospital Tübingen, Tübingen, Germany
| | - Lijuan Cheng
- Department of Hematology, Oncology, Immunology, Rheumatology and Pulmonology, University Hospital Tübingen, Tübingen, Germany
| | - Andrew Zikic
- Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Houra Loghmani
- Department of Hematology, Oncology, Immunology, Rheumatology and Pulmonology, University Hospital Tübingen, Tübingen, Germany
| | - Ivan Tesakov
- Department of Hematology, Oncology, Immunology, Rheumatology and Pulmonology, University Hospital Tübingen, Tübingen, Germany
| | - Patrick Müller
- Friedrich Miescher Laboratory of the Max Planck Society, Tübingen, Germany
| | - Meinolf Suttorp
- Pediatric Hematology and Oncology, Medical Faculty, Technical University Dresden, Dresden, Germany
| | - Karl Welte
- Department of Hematology, Oncology, Immunology, Rheumatology and Pulmonology, University Hospital Tübingen, Tübingen, Germany; Department of Pediatric Hematology, Oncology and Bone Marrow Transplantation, University Hospital Tübingen, Tübingen, Germany
| | - Julia Skokowa
- Department of Hematology, Oncology, Immunology, Rheumatology and Pulmonology, University Hospital Tübingen, Tübingen, Germany
| | - Tatsuya Morishima
- Department of Hematology, Oncology, Immunology, Rheumatology and Pulmonology, University Hospital Tübingen, Tübingen, Germany.
| |
Collapse
|
2
|
Shah V, Lam HY, Leong CHM, Sakaizawa R, Shah JS, Kumar AP. Epigenetic Control of Redox Pathways in Cancer Progression. Antioxid Redox Signal 2025. [PMID: 39815993 DOI: 10.1089/ars.2023.0465] [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] [Indexed: 01/18/2025]
Abstract
Significance: Growing evidence indicates the importance of redox reactions homeostasis, mediated predominantly by reactive oxygen species (ROS) in influencing the development, differentiation, progression, metastasis, programmed cell death, tumor microenvironment, and therapeutic resistance of cancer. Therefore, reviewing the ROS-linked epigenetic changes in cancer is fundamental to understanding the progression and prevention of cancer. Recent Advances: We review in depth the molecular mechanisms involved in ROS-mediated epigenetic changes that lead to alteration of gene expression by altering DNA, modifying histones, and remodeling chromatin and noncoding RNA. Critical Issues: In cancerous cells, alterations of the gene-expression regulatory elements could be generated by the virtue of imbalance in tumor microenvironment. Various oxidizing agents and mitochondrial electron transport chain are the major pathways that generate ROS. ROS plays a key role in carcinogenesis by activating pro-inflammatory signaling pathways and DNA damage. This loss of ROS-mediated epigenetic regulation of the signaling pathways may promote tumorigenesis. We address all such aspects in this review. Future Directions: Developments in this growing field of epigenetics are expected to contribute to further our understanding of human health and diseases such as cancer and to test the clinical applications of redox-based therapy. Recent studies of the cancer-epigenetic landscape have revealed pervasive deregulation of the epigenetic factors in cancer. Thus, the study of interaction between ROS and epigenetic factors in cancer holds a great promise in the development of effective and targeted treatment modalities. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Vandit Shah
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, India
| | - Hiu Yan Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Charlene Hoi-Mun Leong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Reo Sakaizawa
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jigna S Shah
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, India
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
3
|
Valle-Mendiola A, Rocha-Zavaleta L, Maldonado-Lagunas V, Morelos-Laguna D, Gutiérrez-Hoya A, Weiss-Steider B, Soto-Cruz I. STAT5 Is Necessary for the Metabolic Switch Induced by IL-2 in Cervical Cancer Cell Line SiHa. Int J Mol Sci 2024; 25:6835. [PMID: 38999946 PMCID: PMC11241652 DOI: 10.3390/ijms25136835] [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: 04/09/2024] [Revised: 05/07/2024] [Accepted: 06/10/2024] [Indexed: 07/14/2024] Open
Abstract
The tumor cells reprogram their metabolism to cover their high bioenergetic demands for maintaining uncontrolled growth. This response can be mediated by cytokines such as IL-2, which binds to its receptor and activates the JAK/STAT pathway. Some reports show a correlation between the JAK/STAT pathway and cellular metabolism, since the constitutive activation of STAT proteins promotes glycolysis through the transcriptional activation of genes related to energetic metabolism. However, the role of STAT proteins in the metabolic switch induced by cytokines in cervical cancer remains poorly understood. In this study, we analyzed the effect of IL-2 on the metabolic switch and the role of STAT5 in this response. Our results show that IL-2 induces cervical cancer cell proliferation and the tyrosine phosphorylation of STAT5. Also, it induces an increase in lactate secretion and the ratio of NAD+/NADH, which suggest a metabolic reprogramming of their metabolism. When STAT5 was silenced, the lactate secretion and the NAD+/NADH ratio decreased. Also, the expression of HIF1α and GLUT1 decreased. These results indicate that STAT5 regulates IL-2-induced cell proliferation and the metabolic shift to aerobic glycolysis by regulating genes related to energy metabolism. Our results suggest that STAT proteins modulate the metabolic switch in cervical cancer cells to attend to their high demand of energy required for cell growth and proliferation.
Collapse
Affiliation(s)
- Arturo Valle-Mendiola
- Laboratorio de Oncología Molecular, Unidad de Investigación en Diferenciación Celular y Cáncer, FES Zaragoza, Universidad Nacional Autónoma de México, Batalla 5 de Mayo s/n Col. Ejército de Oriente, Mexico City 09230, Mexico; (A.V.-M.); (D.M.-L.); (A.G.-H.); (B.W.-S.)
| | - Leticia Rocha-Zavaleta
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| | - Vilma Maldonado-Lagunas
- Laboratorio de Epigenética, Instituto Nacional de Medicina Genómica (INMEGEN), Periférico Sur no. 4809, Col. Arenal Tepepan, Tlalpan, Mexico City 14610, Mexico;
| | - Diego Morelos-Laguna
- Laboratorio de Oncología Molecular, Unidad de Investigación en Diferenciación Celular y Cáncer, FES Zaragoza, Universidad Nacional Autónoma de México, Batalla 5 de Mayo s/n Col. Ejército de Oriente, Mexico City 09230, Mexico; (A.V.-M.); (D.M.-L.); (A.G.-H.); (B.W.-S.)
| | - Adriana Gutiérrez-Hoya
- Laboratorio de Oncología Molecular, Unidad de Investigación en Diferenciación Celular y Cáncer, FES Zaragoza, Universidad Nacional Autónoma de México, Batalla 5 de Mayo s/n Col. Ejército de Oriente, Mexico City 09230, Mexico; (A.V.-M.); (D.M.-L.); (A.G.-H.); (B.W.-S.)
- Cátedra CONAHCYT, FES Zaragoza, Universidad Nacional Autónoma de México, Mexico City 68020, Mexico
| | - Benny Weiss-Steider
- Laboratorio de Oncología Molecular, Unidad de Investigación en Diferenciación Celular y Cáncer, FES Zaragoza, Universidad Nacional Autónoma de México, Batalla 5 de Mayo s/n Col. Ejército de Oriente, Mexico City 09230, Mexico; (A.V.-M.); (D.M.-L.); (A.G.-H.); (B.W.-S.)
| | - Isabel Soto-Cruz
- Laboratorio de Oncología Molecular, Unidad de Investigación en Diferenciación Celular y Cáncer, FES Zaragoza, Universidad Nacional Autónoma de México, Batalla 5 de Mayo s/n Col. Ejército de Oriente, Mexico City 09230, Mexico; (A.V.-M.); (D.M.-L.); (A.G.-H.); (B.W.-S.)
| |
Collapse
|
4
|
Ponzone L, Audrito V, Landi C, Moiso E, Levra Levron C, Ferrua S, Savino A, Vitale N, Gasparrini M, Avalle L, Vantaggiato L, Shaba E, Tassone B, Saoncella S, Orso F, Viavattene D, Marina E, Fiorilla I, Burrone G, Abili Y, Altruda F, Bini L, Deaglio S, Defilippi P, Menga A, Poli V, Porporato PE, Provero P, Raffaelli N, Riganti C, Taverna D, Cavallo F, Calautti E. RICTOR/mTORC2 downregulation in BRAF V600E melanoma cells promotes resistance to BRAF/MEK inhibition. Mol Cancer 2024; 23:105. [PMID: 38755661 PMCID: PMC11097536 DOI: 10.1186/s12943-024-02010-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 04/26/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND The main drawback of BRAF/MEK inhibitors (BRAF/MEKi)-based targeted therapy in the management of BRAF-mutated cutaneous metastatic melanoma (MM) is the development of therapeutic resistance. We aimed to assess in this context the role of mTORC2, a signaling complex defined by the presence of the essential RICTOR subunit, regarded as an oncogenic driver in several tumor types, including MM. METHODS After analyzing The Cancer Genome Atlas MM patients' database to explore both overall survival and molecular signatures as a function of intra-tumor RICTOR levels, we investigated the effects of RICTOR downregulation in BRAFV600E MM cell lines on their response to BRAF/MEKi. We performed proteomic screening to identify proteins modulated by changes in RICTOR expression, and Seahorse analysis to evaluate the effects of RICTOR depletion on mitochondrial respiration. The combination of BRAFi with drugs targeting proteins and processes emerged in the proteomic screening was carried out on RICTOR-deficient cells in vitro and in a xenograft setting in vivo. RESULTS Low RICTOR levels in BRAF-mutated MM correlate with a worse clinical outcome. Gene Set Enrichment Analysis of low-RICTOR tumors display gene signatures suggestive of activation of the mitochondrial Electron Transport Chain (ETC) energy production. RICTOR-deficient BRAFV600E cells are intrinsically tolerant to BRAF/MEKi and anticipate the onset of resistance to BRAFi upon prolonged drug exposure. Moreover, in drug-naïve cells we observed a decline in RICTOR expression shortly after BRAFi exposure. In RICTOR-depleted cells, both mitochondrial respiration and expression of nicotinamide phosphoribosyltransferase (NAMPT) are enhanced, and their pharmacological inhibition restores sensitivity to BRAFi. CONCLUSIONS Our work unveils an unforeseen tumor-suppressing role for mTORC2 in the early adaptation phase of BRAFV600E melanoma cells to targeted therapy and identifies the NAMPT-ETC axis as a potential therapeutic vulnerability of low RICTOR tumors. Importantly, our findings indicate that the evaluation of intra-tumor RICTOR levels has a prognostic value in metastatic melanoma and may help to guide therapeutic strategies in a personalized manner.
Collapse
Affiliation(s)
- Luca Ponzone
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
| | - Valentina Audrito
- Department of Science and Technological Innovation, University of Piemonte Orientale, Alessandria, 15121, Italy
| | - Claudia Landi
- Functional Proteomic Section, Department of Life Sciences, University of Siena, Siena, 53100, Italy
| | - Enrico Moiso
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Chiara Levra Levron
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Life Sciences and Systems Biology, University of Turin, Turin, 10126, Italy
| | - Sara Ferrua
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
| | - Aurora Savino
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
| | - Nicoletta Vitale
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
| | - Massimiliano Gasparrini
- Department of Agriculture, Food and Environmental Sciences, Polytechnic University of Marche, Ancona, 60131, Italy
| | - Lidia Avalle
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
- Department of Science and Technological Innovation, University of Piemonte Orientale, Alessandria, 15121, Italy
| | - Lorenza Vantaggiato
- Functional Proteomic Section, Department of Life Sciences, University of Siena, Siena, 53100, Italy
| | - Enxhi Shaba
- Functional Proteomic Section, Department of Life Sciences, University of Siena, Siena, 53100, Italy
| | - Beatrice Tassone
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
- Department of Personal Care, dsm-firmenich, Kaiseraugst, 4303, Switzerland
| | - Stefania Saoncella
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
| | - Francesca Orso
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
| | - Daniele Viavattene
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
| | - Eleonora Marina
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
| | - Irene Fiorilla
- Department of Science and Technological Innovation, University of Piemonte Orientale, Alessandria, 15121, Italy
| | - Giulia Burrone
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
- Department of Clinical and Biological Sciences, University of Turin, Turin, 10124, Italy
| | - Youssef Abili
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
- GenomeUp, Rome, 00144, Italy
| | - Fiorella Altruda
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
| | - Luca Bini
- Functional Proteomic Section, Department of Life Sciences, University of Siena, Siena, 53100, Italy
| | - Silvia Deaglio
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Medical Sciences, University of Turin, Turin, 10124, Italy
| | - Paola Defilippi
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
| | - Alessio Menga
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
| | - Valeria Poli
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
| | - Paolo Ettore Porporato
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
| | - Paolo Provero
- Neuroscience Department "Rita Levi Montalcini", University of Turin, Turin, 10126, Italy
| | - Nadia Raffaelli
- Department of Agriculture, Food and Environmental Sciences, Polytechnic University of Marche, Ancona, 60131, Italy
| | - Chiara Riganti
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Oncology, University of Turin, Turin, 10124, Italy
| | - Daniela Taverna
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
| | - Federica Cavallo
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
| | - Enzo Calautti
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy.
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy.
| |
Collapse
|
5
|
Proteau S, Krossa I, Husser C, Guéguinou M, Sella F, Bille K, Irondelle M, Dalmasso M, Barouillet T, Cheli Y, Pisibon C, Arrighi N, Nahon‐Estève S, Martel A, Gastaud L, Lassalle S, Mignen O, Brest P, Mazure NM, Bost F, Baillif S, Landreville S, Turcotte S, Hasson D, Carcamo S, Vandier C, Bernstein E, Yvan‐Charvet L, Levesque MP, Ballotti R, Bertolotto C, Strub T. LKB1-SIK2 loss drives uveal melanoma proliferation and hypersensitivity to SLC8A1 and ROS inhibition. EMBO Mol Med 2023; 15:e17719. [PMID: 37966164 PMCID: PMC10701601 DOI: 10.15252/emmm.202317719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 10/16/2023] [Accepted: 10/23/2023] [Indexed: 11/16/2023] Open
Abstract
Metastatic uveal melanomas are highly resistant to all existing treatments. To address this critical issue, we performed a kinome-wide CRISPR-Cas9 knockout screen, which revealed the LKB1-SIK2 module in restraining uveal melanoma tumorigenesis. Functionally, LKB1 loss enhances proliferation and survival through SIK2 inhibition and upregulation of the sodium/calcium (Na+ /Ca2+ ) exchanger SLC8A1. This signaling cascade promotes increased levels of intracellular calcium and mitochondrial reactive oxygen species, two hallmarks of cancer. We further demonstrate that combination of an SLC8A1 inhibitor and a mitochondria-targeted antioxidant promotes enhanced cell death efficacy in LKB1- and SIK2-negative uveal melanoma cells compared to control cells. Our study also identified an LKB1-loss gene signature for the survival prognostic of patients with uveal melanoma that may be also predictive of response to the therapy combination. Our data thus identify not only metabolic vulnerabilities but also new prognostic markers, thereby providing a therapeutic strategy for particular subtypes of metastatic uveal melanoma.
Collapse
Affiliation(s)
- Sarah Proteau
- University Côte d'AzurNiceFrance
- Inserm, Biology and Pathologies of melanocytes, team1, Equipe labellisée Ligue 2020, and Equipe labellisée ARC 2022, Mediterranean Centre for Molecular MedicineNiceFrance
| | - Imène Krossa
- University Côte d'AzurNiceFrance
- Inserm, Biology and Pathologies of melanocytes, team1, Equipe labellisée Ligue 2020, and Equipe labellisée ARC 2022, Mediterranean Centre for Molecular MedicineNiceFrance
| | - Chrystel Husser
- University Côte d'AzurNiceFrance
- Inserm, Biology and Pathologies of melanocytes, team1, Equipe labellisée Ligue 2020, and Equipe labellisée ARC 2022, Mediterranean Centre for Molecular MedicineNiceFrance
| | | | - Federica Sella
- Department of Dermatology, University Hospital ZurichUniversity of ZurichZurichSwitzerland
| | - Karine Bille
- University Côte d'AzurNiceFrance
- Inserm, Biology and Pathologies of melanocytes, team1, Equipe labellisée Ligue 2020, and Equipe labellisée ARC 2022, Mediterranean Centre for Molecular MedicineNiceFrance
| | | | - Mélanie Dalmasso
- University Côte d'AzurNiceFrance
- Inserm, Biology and Pathologies of melanocytes, team1, Equipe labellisée Ligue 2020, and Equipe labellisée ARC 2022, Mediterranean Centre for Molecular MedicineNiceFrance
| | - Thibault Barouillet
- Inserm, Hematometabolism and metainflammation, team 13, Mediterranean Centre for Molecular MedicineNiceFrance
| | - Yann Cheli
- University Côte d'AzurNiceFrance
- Inserm, Biology and Pathologies of melanocytes, team1, Equipe labellisée Ligue 2020, and Equipe labellisée ARC 2022, Mediterranean Centre for Molecular MedicineNiceFrance
| | - Céline Pisibon
- University Côte d'AzurNiceFrance
- Inserm, Biology and Pathologies of melanocytes, team1, Equipe labellisée Ligue 2020, and Equipe labellisée ARC 2022, Mediterranean Centre for Molecular MedicineNiceFrance
| | - Nicole Arrighi
- University Côte d'AzurNiceFrance
- Inserm, Biology and Pathologies of melanocytes, team1, Equipe labellisée Ligue 2020, and Equipe labellisée ARC 2022, Mediterranean Centre for Molecular MedicineNiceFrance
| | - Sacha Nahon‐Estève
- University Côte d'AzurNiceFrance
- Department of OphthalmologyCentre Hospitalier Universitaire of NiceNiceFrance
| | - Arnaud Martel
- University Côte d'AzurNiceFrance
- Department of OphthalmologyCentre Hospitalier Universitaire of NiceNiceFrance
| | | | - Sandra Lassalle
- University Côte d'AzurNiceFrance
- Laboratory of Clinical and Experimental Pathology, University Hospital of Nice, FHU OncoAge, Cote d'Azur University, Biobank BB‐0033‐00025, IRCAN team 4, OncoAge FHUNiceFrance
| | | | - Patrick Brest
- University Côte d'AzurNiceFrance
- IRCAN team 4, Inserm, CNRS, FHU‐oncoAge, IHU‐RESPIRera NiceNiceFrance
| | - Nathalie M Mazure
- University Côte d'AzurNiceFrance
- Inserm, Cancer, Metabolism and environment, team, Equipe labellisée Ligue 2022, Mediterranean Centre for Molecular MedicineNiceFrance
| | - Frédéric Bost
- University Côte d'AzurNiceFrance
- Inserm, Cancer, Metabolism and environment, team, Equipe labellisée Ligue 2022, Mediterranean Centre for Molecular MedicineNiceFrance
| | - Stéphanie Baillif
- University Côte d'AzurNiceFrance
- Department of OphthalmologyCentre Hospitalier Universitaire of NiceNiceFrance
| | - Solange Landreville
- Département d'ophtalmologie et d'ORL‐CCF, Faculté de médecineUniversité LavalQuebec CityQCCanada
- CUO‐Recherche and Axe médecine régénératriceCentre de recherche du CHU de Québec‐Université LavalQuebec CityQCCanada
- Centre de recherche sur le cancer de l'Université LavalQuebec CityQCCanada
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEXQuebec CityQCCanada
| | - Simon Turcotte
- Cancer AxisCentre de recherche du Centre Hospitalier de l'Université de Montréal/Institut du cancer de MontréalMontréalQCCanada
- Hepato‐Pancreato‐Biliary Surgery and Liver Transplantation ServiceCentre hospitalier de l'Université de MontréalMontréalQCCanada
| | - Dan Hasson
- Department of Oncological Sciences, Tisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNYUSA
- Tisch Cancer Institute Bioinformatics for Next Generation Sequencing (BiNGS) FacilityIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Saul Carcamo
- Department of Oncological Sciences, Tisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNYUSA
- Tisch Cancer Institute Bioinformatics for Next Generation Sequencing (BiNGS) FacilityIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | | | - Emily Bernstein
- Department of Oncological Sciences, Tisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Laurent Yvan‐Charvet
- University Côte d'AzurNiceFrance
- Inserm, Hematometabolism and metainflammation, team 13, Mediterranean Centre for Molecular MedicineNiceFrance
| | - Mitchell P Levesque
- Department of Dermatology, University Hospital ZurichUniversity of ZurichZurichSwitzerland
| | - Robert Ballotti
- University Côte d'AzurNiceFrance
- Inserm, Biology and Pathologies of melanocytes, team1, Equipe labellisée Ligue 2020, and Equipe labellisée ARC 2022, Mediterranean Centre for Molecular MedicineNiceFrance
| | - Corine Bertolotto
- University Côte d'AzurNiceFrance
- Inserm, Biology and Pathologies of melanocytes, team1, Equipe labellisée Ligue 2020, and Equipe labellisée ARC 2022, Mediterranean Centre for Molecular MedicineNiceFrance
| | - Thomas Strub
- University Côte d'AzurNiceFrance
- Inserm, Biology and Pathologies of melanocytes, team1, Equipe labellisée Ligue 2020, and Equipe labellisée ARC 2022, Mediterranean Centre for Molecular MedicineNiceFrance
| |
Collapse
|
6
|
Yong J, Cai S, Zeng Z. Targeting NAD + metabolism: dual roles in cancer treatment. Front Immunol 2023; 14:1269896. [PMID: 38116009 PMCID: PMC10728650 DOI: 10.3389/fimmu.2023.1269896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 11/20/2023] [Indexed: 12/21/2023] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) is indispensable for various oxidation-reduction reactions in mammalian cells, particularly during energy production. Malignant cells increase the expression levels of NAD+ biosynthesis enzymes for rapid proliferation and biomass production. Furthermore, mounting proof has indicated that NAD-degrading enzymes (NADases) play a role in creating the immunosuppressive tumor microenvironment (TME). Interestingly, both inhibiting NAD+ synthesis and targeting NADase have positive implications for cancer treatment. Here we summarize the detrimental outcomes of increased NAD+ production, the functions of NAD+ metabolic enzymes in creating an immunosuppressive TME, and discuss the progress and clinical translational potential of inhibitors for NAD+ synthesis and therapies targeting NADase.
Collapse
Affiliation(s)
- Jiaxin Yong
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Songqing Cai
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Zhaolei Zeng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
7
|
Molinelli E, Ceccarelli G, Fantone S, Di Mercurio E, Gambini D, Maurizi A, Perugini J, Tossetta G, Brisigotti V, De Simoni E, Sapigni C, Rizzetto G, Campanati A, Simonetti O, Marzioni D, Offidani A. Melanoma and subcutaneous adipose tissue: Role of peritumoral adipokines in disease characterization and prognosis. Pigment Cell Melanoma Res 2023; 36:423-430. [PMID: 37334675 DOI: 10.1111/pcmr.13103] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 05/08/2023] [Accepted: 05/26/2023] [Indexed: 06/20/2023]
Abstract
In the last decades, the concept of adipose organ has emerged, giving adipose tissue an active endocrine and immunologic function through the secretion of multiple cytokines and chemokines that seem to be implicated in the development and progression of several cancer, including cutaneous melanoma. In this pilot experimental study, we analyzed the expression in the peritumor subcutaneous adipose tissue of the most significant adipokines involved in the processes of carcinogenesis and metastasis in a population of melanoma patients and in two control groups composed of melanocytic nevi and epidermoid cysts, respectively. We correlated the results obtained with the main disease prognostic factors observing a statistically significant increase in the expression of PAI1, LEP, CXCL1, NAMPT, and TNF-α at the level of the peritumor tissue of the melanoma samples compared to the control groups and a correlation of the same with the histopathological prognostic factor of melanoma. Our preliminary study shows that the overexpression of PAI1, LEP, CXCL1, NAMPT, and TNF-α may contribute to the growth and to the local aggressiveness of cutaneous melanoma. It opens the hypothesis of a direct oncogenic role of subcutaneous adipose tissue and adipokines in the tumorigenesis of melanoma.
Collapse
Affiliation(s)
- Elisa Molinelli
- Dermatological Unit, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | | | - Sonia Fantone
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Eleonora Di Mercurio
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Daisy Gambini
- Dermatological Unit, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Andrea Maurizi
- Dermatological Unit, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Jessica Perugini
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Giovanni Tossetta
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Valerio Brisigotti
- Dermatological Unit, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Edoardo De Simoni
- Dermatological Unit, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Claudia Sapigni
- Dermatological Unit, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Giulio Rizzetto
- Dermatological Unit, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Anna Campanati
- Dermatological Unit, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Oriana Simonetti
- Dermatological Unit, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Daniela Marzioni
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Annamaria Offidani
- Dermatological Unit, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
8
|
Yang X, Wang X, Yang Y, Li Z, Chen Y, Shang S, Wang Y. DNMT3A mutation promotes leukemia development through NAM-NAD metabolic reprogramming. J Transl Med 2023; 21:481. [PMID: 37464424 PMCID: PMC10355022 DOI: 10.1186/s12967-023-04323-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 07/01/2023] [Indexed: 07/20/2023] Open
Abstract
BACKGROUND DNA methyltransferase 3A (DNMT3A) is frequently mutated in acute myeloid leukemia (AML) with Arg882His (R882H) as the hotspot mutation. It has been reported that DNMT3A mutation plays a key role in leukemogenesis through hypomethylation of some target genes associated with cell growth and differentiation. In this study, we investigated the function of DNMT3A R882H in the malignant progression of AML by regulating metabolic reprogramming. METHODS Ultra-High Performance Liquid Chromatography-High Resolution Tandem Mass Spectrometry (UHPLC-HRMS/MS) was used to detect metabolites in the serum of mice harboring Dnmt3a R878H mutation and the wild-type Dnmt3a. Methylated DNA Immunoprecipitation Sequencing (MeDIP-seq) and RNA sequencing (RNA-seq) were used to analyze the levels of DNA methylation and mRNA expression of genes in mouse Gr1+ bone marrow cells respectively. The TCGA and GO databases were used to analyze the differential genes between human samples carrying the DNMT3A R882 mutation and the wild-type DNMT3A. Co-immunoprecipitation and immunoblotting were used to illustrate the binding levels of Cyclins-CDKs and CDK inhibitors including CDKN1A and CDKN1B. Flow cytometry was used to analyze the cell differentiation, division, apoptosis and cell cycle. The effect of NAMPT inhibition on leukemia was evaluated by using in vivo fluorescence imaging in NOG mouse model bearing OCI-AML3 cells. RESULTS DNMT3A mutation caused high expression of nicotinamide phosphoribosyltransferase (NAMPT), a key enzyme in the nicotinamide adenine dinucleotide (NAD) salvage synthetic pathway, through DNA hypomethylation, and finally led to abnormal nicotinamide (NAM) metabolism and NAD synthesis. The NAM-NAD metabolic abnormalities caused accelerated cell cycle progression. Inhibition of NAMPT can reduce the binding degree between Cyclins-CDKs, and increase the binding interaction of the CDK inhibitors with Cyclins-CDKs complexes. Moreover, cells with high expression of NAMPT were more sensitive to the NAMPT inhibitor FK866 with a lower IC50. The inhibition of NAMPT can remarkably extend the survival time of tumor-bearing mice and reduce the infiltration of tumor cells. CONCLUSIONS Taken together, our data showed that DNMT3A mutation caused NAMPT overexpression to induce the reprogramming of NAM-NAD metabolism and contribute to abnormal proliferation, which provided a potential direction for targeted therapy at the metabolic level in AML with DNMT3A mutation.
Collapse
Affiliation(s)
- Xuejiao Yang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiao Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ying Yang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zhiyang Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430000, Hubei, China
| | - Yunshuo Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Siqi Shang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yueying Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
9
|
Tang H, Wang L, Wang T, Yang J, Zheng S, Tong J, Jiang S, Zhang X, Zhang K. Recent advances of targeting nicotinamide phosphoribosyltransferase (NAMPT) for cancer drug discovery. Eur J Med Chem 2023; 258:115607. [PMID: 37413882 DOI: 10.1016/j.ejmech.2023.115607] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/16/2023] [Accepted: 06/26/2023] [Indexed: 07/08/2023]
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) is the rate-limiting enzyme for the biosynthesis of NAD+ in the salvage pathway. NAMPT is overexpressed in various cancers, associating with a poor prognosis and tumor progression. Beyond cancer metabolism, recent evidence unravels additional roles of NAMPT in cancer biology, including DNA repair machinery, crosstalk with oncogenic signaling pathways, cancer cell stemness, and immune responses. NAMPT is a promising therapeutic target for cancer. However, first-generation NAMPT inhibitors exhibited limited efficacy and dose-limiting toxicities in clinical trials. Multiple strategies are being exploited to improve their efficacy and minimize toxic-side effects. This review discusses the biomarkers predictive of response to NAMPT inhibitors, and summarizes the most significant advances in the evolution of structurally distinct NAMPT inhibitors, the manipulation of targeted delivery technologies via antibody-drug conjugates (ADCs), PhotoActivated ChemoTherapy (PACT) and the intratumoral delivery system, as well as the development and pharmacological outcomes of NAMPT degraders. Finally, a discussion of future perspectives and challenges in this area is also included.
Collapse
Affiliation(s)
- He Tang
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Lin Wang
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Tianyu Wang
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Jiamei Yang
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Shuai Zheng
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Jun Tong
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Sheng Jiang
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Xiangyu Zhang
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Kuojun Zhang
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
10
|
Abstract
Over the past decade, melanoma has led the field in new cancer treatments, with impressive gains in on-treatment survival but more modest improvements in overall survival. Melanoma presents heterogeneity and transcriptional plasticity that recapitulates distinct melanocyte developmental states and phenotypes, allowing it to adapt to and eventually escape even the most advanced treatments. Despite remarkable advances in our understanding of melanoma biology and genetics, the melanoma cell of origin is still fiercely debated because both melanocyte stem cells and mature melanocytes can be transformed. Animal models and high-throughput single-cell sequencing approaches have opened new opportunities to address this question. Here, we discuss the melanocytic journey from the neural crest, where they emerge as melanoblasts, to the fully mature pigmented melanocytes resident in several tissues. We describe a new understanding of melanocyte biology and the different melanocyte subpopulations and microenvironments they inhabit, and how this provides unique insights into melanoma initiation and progression. We highlight recent findings on melanoma heterogeneity and transcriptional plasticity and their implications for exciting new research areas and treatment opportunities. The lessons from melanocyte biology reveal how cells that are present to protect us from the damaging effects of ultraviolet radiation reach back to their origins to become a potentially deadly cancer.
Collapse
Affiliation(s)
- Patricia P Centeno
- Molecular Oncology Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, UK
| | - Valeria Pavet
- Molecular Oncology Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, UK
| | - Richard Marais
- Molecular Oncology Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, UK.
- Oncodrug Ltd, Alderly Park, Macclesfield, UK.
| |
Collapse
|
11
|
Rempuia V, Gurusubramanian G, Roy VK. Evidence of the inhibitory role of visfatin in the testicular activity of mice during the infantile stage. J Steroid Biochem Mol Biol 2023; 231:106306. [PMID: 37024018 DOI: 10.1016/j.jsbmb.2023.106306] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 02/07/2023] [Accepted: 04/03/2023] [Indexed: 04/08/2023]
Abstract
Adipokines have emerged as regulators of gonadal function in many mammalian and non-mammalian species. In the present study, we have investigated the developmental expression of testicular and ovarian visfatin along with its possible role in the testicular activity infantile stages. Previously, our group has the extensive role of ovarian visfatin in relation to steroidogenesis, proliferation, and apoptosis in female mice. To the best of our knowledge, no study has shown the role of visfatin in mice testis. Our results from the previous study and present study showed that visfatin in the testis and ovaries are developmentally regulated. To unravel the role of visfatin, we have used FK866, as visfatin inhibitor. FK866 was used as a visfatin inhibitor, to decipher the role of visfatin in the testis of mice. Our results showed that visfatin expression in the testis was developmentally regulated in the testis. Leydig cells as well as germ have shown the presence of visfatin in mice testis, which suggest its role in testicular steroidogenesis and spermatogenesis. Furthermore, visfatin inhibition by FK866 significantly increased the testosterone secretion, and expression of AR, Bcl2, and ERα. The expression of GCNA was upregulated by FK866 treatment. These results suggest that visfatin has an inhibitory role in testicular steroidogenesis and germ cell proliferation in the infantile stage. Further research is required to define the precise role of visfatin in infantile mice testis.
Collapse
Affiliation(s)
- Vanlal Rempuia
- Department of Zoology, Mizoram University, Aizawl-796004, Mizoram, India
| | | | - Vikas Kumar Roy
- Department of Zoology, Mizoram University, Aizawl-796004, Mizoram, India.
| |
Collapse
|
12
|
Li YJ, Zhang C, Martincuks A, Herrmann A, Yu H. STAT proteins in cancer: orchestration of metabolism. Nat Rev Cancer 2023; 23:115-134. [PMID: 36596870 DOI: 10.1038/s41568-022-00537-3] [Citation(s) in RCA: 123] [Impact Index Per Article: 61.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/14/2022] [Indexed: 01/04/2023]
Abstract
Reprogrammed metabolism is a hallmark of cancer. However, the metabolic dependency of cancer, from tumour initiation through disease progression and therapy resistance, requires a spectrum of distinct reprogrammed cellular metabolic pathways. These pathways include aerobic glycolysis, oxidative phosphorylation, reactive oxygen species generation, de novo lipid synthesis, fatty acid β-oxidation, amino acid (notably glutamine) metabolism and mitochondrial metabolism. This Review highlights the central roles of signal transducer and activator of transcription (STAT) proteins, notably STAT3, STAT5, STAT6 and STAT1, in orchestrating the highly dynamic metabolism not only of cancer cells but also of immune cells and adipocytes in the tumour microenvironment. STAT proteins are able to shape distinct metabolic processes that regulate tumour progression and therapy resistance by transducing signals from metabolites, cytokines, growth factors and their receptors; defining genetic programmes that regulate a wide range of molecules involved in orchestration of metabolism in cancer and immune cells; and regulating mitochondrial activity at multiple levels, including energy metabolism and lipid-mediated mitochondrial integrity. Given the central role of STAT proteins in regulation of metabolic states, they are potential therapeutic targets for altering metabolic reprogramming in cancer.
Collapse
Affiliation(s)
- Yi-Jia Li
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Chunyan Zhang
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Antons Martincuks
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Andreas Herrmann
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
- Sorrento Therapeutics, San Diego, CA, USA
| | - Hua Yu
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA.
| |
Collapse
|
13
|
Barba C, Ekiz HA, Tang WW, Ghazaryan A, Hansen M, Lee SH, Voth WP, O’Connell RM. Interferon Gamma-Inducible NAMPT in Melanoma Cells Serves as a Mechanism of Resistance to Enhance Tumor Growth. Cancers (Basel) 2023; 15:1411. [PMID: 36900204 PMCID: PMC10000695 DOI: 10.3390/cancers15051411] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/15/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
(1) Background: Immune cells infiltrate the tumor microenvironment and secrete inflammatory cytokines, including interferons (IFNs), to drive antitumor responses and promote tumor clearance. However, recent evidence suggests that sometimes, tumor cells can also harness IFNs to enhance growth and survival. The essential NAD+ salvage pathway enzyme nicotinamide phosphoribosyltransferase (NAMPT) gene is constitutively expressed in cells during normal homeostasis. However, melanoma cells have higher energetic demands and elevated NAMPT expression. We hypothesized that interferon gamma (IFNγ) regulates NAMPT in tumor cells as a mechanism of resistance that impedes the normal anti-tumorigenic effects of IFNγ. (2) Methods: Utilizing a variety of melanoma cells, mouse models, Crispr-Cas9, and molecular biology techniques, we explored the importance of IFNγ-inducible NAMPT during melanoma growth. (3) Results: We demonstrated that IFNγ mediates the metabolic reprogramming of melanoma cells by inducing Nampt through a Stat1 binding site in the Nampt gene, increasing cell proliferation and survival. Further, IFN/STAT1-inducible Nampt promotes melanoma in vivo. (4) Conclusions: We provided evidence that melanoma cells directly respond to IFNγ by increasing NAMPT levels, improving their fitness and growth in vivo (control n = 36, SBS KO n = 46). This discovery unveils a possible therapeutic target that may improve the efficacy of immunotherapies involving IFN responses in the clinic.
Collapse
Affiliation(s)
- Cindy Barba
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| | - H. Atakan Ekiz
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
- Izmir Institute of Technology, Molecular Biology and Genetics Department, Gulbahce, Izmir 35430, Turkey
| | - William Weihao Tang
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| | - Arevik Ghazaryan
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| | - Mason Hansen
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| | - Soh-Hyun Lee
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| | - Warren Peter Voth
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| | - Ryan Michael O’Connell
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
14
|
Ge T, Gu X, Jia R, Ge S, Chai P, Zhuang A, Fan X. Crosstalk between metabolic reprogramming and epigenetics in cancer: updates on mechanisms and therapeutic opportunities. CANCER COMMUNICATIONS (LONDON, ENGLAND) 2022; 42:1049-1082. [PMID: 36266736 PMCID: PMC9648395 DOI: 10.1002/cac2.12374] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 09/19/2022] [Accepted: 10/10/2022] [Indexed: 11/24/2022]
Abstract
Reversible, spatial, and temporal regulation of metabolic reprogramming and epigenetic homeostasis are prominent hallmarks of carcinogenesis. Cancer cells reprogram their metabolism to meet the high bioenergetic and biosynthetic demands for vigorous proliferation. Epigenetic dysregulation is a common feature of human cancers, which contributes to tumorigenesis and maintenance of the malignant phenotypes by regulating gene expression. The epigenome is sensitive to metabolic changes. Metabolism produces various metabolites that are substrates, cofactors, or inhibitors of epigenetic enzymes. Alterations in metabolic pathways and fluctuations in intermediate metabolites convey information regarding the intracellular metabolic status into the nucleus by modulating the activity of epigenetic enzymes and thus remodeling the epigenetic landscape, inducing transcriptional responses to heterogeneous metabolic requirements. Cancer metabolism is regulated by epigenetic machinery at both transcriptional and post‐transcriptional levels. Epigenetic modifiers, chromatin remodelers and non‐coding RNAs are integral contributors to the regulatory networks involved in cancer metabolism, facilitating malignant transformation. However, the significance of the close connection between metabolism and epigenetics in the context of cancer has not been fully deciphered. Thus, it will be constructive to summarize and update the emerging new evidence supporting this bidirectional crosstalk and deeply assess how the crosstalk between metabolic reprogramming and epigenetic abnormalities could be exploited to optimize treatment paradigms and establish new therapeutic options. In this review, we summarize the central mechanisms by which epigenetics and metabolism reciprocally modulate each other in cancer and elaborate upon and update the major contributions of the interplays between epigenetic aberrations and metabolic rewiring to cancer initiation and development. Finally, we highlight the potential therapeutic opportunities for hematological malignancies and solid tumors by targeting this epigenetic‐metabolic circuit. In summary, we endeavored to depict the current understanding of the coordination between these fundamental abnormalities more comprehensively and provide new perspectives for utilizing metabolic and epigenetic targets for cancer treatment.
Collapse
Affiliation(s)
- Tongxin Ge
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, P. R. China
| | - Xiang Gu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, P. R. China
| | - Renbing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, P. R. China
| | - Shengfang Ge
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, P. R. China
| | - Peiwei Chai
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, P. R. China
| | - Ai Zhuang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, P. R. China
| | - Xianqun Fan
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, P. R. China
| |
Collapse
|
15
|
Wei Y, Xiang H, Zhang W. Review of various NAMPT inhibitors for the treatment of cancer. Front Pharmacol 2022; 13:970553. [PMID: 36160449 PMCID: PMC9490061 DOI: 10.3389/fphar.2022.970553] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) is a rate-limiting enzyme in the NAD salvage pathway of mammalian cells and is overexpressed in numerous types of cancers. These include breast cancer, ovarian cancer, prostate cancer, gastric cancer, colorectal cancer, glioma, and b-cell lymphoma. NAMPT is also known to impact the NAD and NADPH pool. Research has demonstrated that NAMPT can be inhibited. NAMPT inhibitors are diverse anticancer medicines with significant anti-tumor efficacy in ex vivo tumor models. A few notable NAMPT specific inhibitors which have been produced include FK866, CHS828, and OT-82. Despite encouraging preclinical evidence of the potential utility of NAMPT inhibitors in cancer models, early clinical trials have yielded only modest results, necessitating the adaptation of additional tactics to boost efficacy. This paper examines a number of cancer treatment methods which target NAMPT, including the usage of individual inhibitors, pharmacological combinations, dual inhibitors, and ADCs, all of which have demonstrated promising experimental or clinical results. We intend to contribute further ideas regarding the usage and development of NAMPT inhibitors in clinical therapy to advance the field of research on this intriguing target.
Collapse
Affiliation(s)
- Yichen Wei
- West China School of Pharmacy, Sichuan University, Chengdu, China
- State Key Laboratory of Biotherapy and Cancer Center, Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Haotian Xiang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Wenqiu Zhang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Wenqiu Zhang,
| |
Collapse
|
16
|
NAD/NAMPT and mTOR Pathways in Melanoma: Drivers of Drug Resistance and Prospective Therapeutic Targets. Int J Mol Sci 2022; 23:ijms23179985. [PMID: 36077374 PMCID: PMC9456568 DOI: 10.3390/ijms23179985] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
Malignant melanoma represents the most fatal skin cancer due to its aggressive behavior and high metastatic potential. The introduction of BRAF/MEK inhibitors and immune-checkpoint inhibitors (ICIs) in the clinic has dramatically improved patient survival over the last decade. However, many patients either display primary (i.e., innate) or develop secondary (i.e., acquired) resistance to systemic treatments. Therapeutic resistance relies on the rewiring of multiple processes, including cancer metabolism, epigenetics, gene expression, and interactions with the tumor microenvironment that are only partially understood. Therefore, reliable biomarkers of resistance or response, capable of facilitating the choice of the best treatment option for each patient, are currently missing. Recently, activation of nicotinamide adenine dinucleotide (NAD) metabolism and, in particular, of its rate-limiting enzyme nicotinamide phosphoribosyltransferase (NAMPT) have been identified as key drivers of targeted therapy resistance and melanoma progression. Another major player in this context is the mammalian target of rapamycin (mTOR) pathway, which plays key roles in the regulation of melanoma cell anabolic functions and energy metabolism at the switch between sensitivity and resistance to targeted therapy. In this review, we summarize known resistance mechanisms to ICIs and targeted therapy, focusing on metabolic adaptation as one main mechanism of drug resistance. In particular, we highlight the roles of NAD/NAMPT and mTOR signaling axes in this context and overview data in support of their inhibition as a promising strategy to overcome treatment resistance.
Collapse
|
17
|
Ohanna M, Biber P, Deckert M. Emerging Role of Deubiquitinating Enzymes (DUBs) in Melanoma Pathogenesis. Cancers (Basel) 2022; 14:3371. [PMID: 35884430 PMCID: PMC9322030 DOI: 10.3390/cancers14143371] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 02/04/2023] Open
Abstract
Metastatic melanoma is the leading cause of death from skin cancer. Therapies targeting the BRAF oncogenic pathway and immunotherapies show remarkable clinical efficacy. However, these treatments are limited to subgroups of patients and relapse is common. Overall, the majority of patients require additional treatments, justifying the development of new therapeutic strategies. Non-genetic and genetic alterations are considered to be important drivers of cellular adaptation mechanisms to current therapies and disease relapse. Importantly, modification of the overall proteome in response to non-genetic and genetic events supports major cellular changes that are required for the survival, proliferation, and migration of melanoma cells. However, the mechanisms underlying these adaptive responses remain to be investigated. The major contributor to proteome remodeling involves the ubiquitin pathway, ubiquitinating enzymes, and ubiquitin-specific proteases also known as DeUBiquitinases (DUBs). In this review, we summarize the current knowledge regarding the nature and roles of the DUBs recently identified in melanoma progression and therapeutic resistance and discuss their potential as novel sources of vulnerability for melanoma therapy.
Collapse
Affiliation(s)
- Mickael Ohanna
- Université Côte d’Azur, INSERM, C3M, 06204 Nice, France; (P.B.); (M.D.)
- Team MicroCan, Equipe Labellisée Ligue Contre le Cancer, 06204 Nice, France
| | - Pierric Biber
- Université Côte d’Azur, INSERM, C3M, 06204 Nice, France; (P.B.); (M.D.)
- Team MicroCan, Equipe Labellisée Ligue Contre le Cancer, 06204 Nice, France
| | - Marcel Deckert
- Université Côte d’Azur, INSERM, C3M, 06204 Nice, France; (P.B.); (M.D.)
- Team MicroCan, Equipe Labellisée Ligue Contre le Cancer, 06204 Nice, France
| |
Collapse
|
18
|
The Kynurenine Pathway and Cancer: Why Keep It Simple When You Can Make It Complicated. Cancers (Basel) 2022; 14:cancers14112793. [PMID: 35681770 PMCID: PMC9179486 DOI: 10.3390/cancers14112793] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/01/2022] [Accepted: 06/03/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary The kynurenine pathway has two main physiological roles: (i) it protects specific organs such as the eyes and placenta from strong immune reactions and (ii) it additionally generate in the liver and kidney a metabolite essential to all cells of human body. Abnormal activation of this pathway is recurrently observed in numerous cancer types. Its two functions are hijacked to promote tumor growth and cancer cell dissemination through multiple mechanisms. Clinical assays including administration of inhibitors of this pathway have not yet been successful. The complex regulation of this pathway is likely the reason behind this failure. In this review, we try to give an overview of the current knowledge about this pathway, to point out the next challenges, and to propose alternative therapeutic routes. Abstract The kynurenine pathway has been highlighted as a gatekeeper of immune-privileged sites through its ability to generate from tryptophan a set of immunosuppressive metabolic intermediates. It additionally constitutes an important source of cellular NAD+ for the organism. Hijacking of its immunosuppressive functions, as recurrently observed in multiple cancers, facilitates immune evasion and promotes tumor development. Based on these observations, researchers have focused on characterizing indoleamine 2,3-dioxygenase (IDO1), the main enzyme catalyzing the first and limiting step of the pathway, and on developing therapies targeting it. Unfortunately, clinical trials studying IDO1 inhibitors have thus far not met expectations, highlighting the need to unravel this complex signaling pathway further. Recent advances demonstrate that these metabolites additionally promote tumor growth, metastatic dissemination and chemoresistance by a combination of paracrine and autocrine effects. Production of NAD+ also contributes to cancer progression by providing cancer cells with enhanced plasticity, invasive properties and chemoresistance. A comprehensive survey of this complexity is challenging but necessary to achieve medical success.
Collapse
|
19
|
Dahmani C, Corre E, Dandou S, Mangé A, Radulescu O, Coopman PJ, Cuq P, Larive RM. La résistance aux inhibiteurs de BRAF. Med Sci (Paris) 2022; 38:570-578. [DOI: 10.1051/medsci/2022083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
La voie de signalisation MAPK/ERK est une voie centrale de la signalisation intracellulaire. Sa dérégulation participe à la transformation et la progression tumorales. Dans plusieurs cancers, la découverte de mutations activatrices de BRAF, à l’origine de l’activation de cette voie, a ouvert de nouvelles perspectives thérapeutiques avec le développement d’inhibiteurs spécifiques de la protéine. Selon les cancers, ces inhibiteurs ont cependant montré soit une efficacité insuffisante, due à la résistance primaire des cellules tumorales, soit une efficacité transitoire, due à l’apparition d’une résistance acquise. Dans cette revue, nous revenons sur les découvertes qui ont conduit au développement de ces inhibiteurs de BRAF. Nous détaillons également les mécanismes moléculaires et cellulaires de la résistance à ces inhibiteurs observée dans différents types de cancers. Comprendre ces mécanismes est en effet primordial pour développer des stratégies thérapeutiques qui soient plus efficaces.
Collapse
|
20
|
Deng Y, Hu B, Miao Y, Wang J, Zhang S, Wan H, Wu Z, Lv Y, Feng J, Ji N, Park D, Hao S. A Nicotinamide Phosphoribosyltransferase Inhibitor, FK866, Suppresses the Growth of Anaplastic Meningiomas and Inhibits Immune Checkpoint Expression by Regulating STAT1. Front Oncol 2022; 12:836257. [PMID: 35515130 PMCID: PMC9065474 DOI: 10.3389/fonc.2022.836257] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/22/2022] [Indexed: 11/18/2022] Open
Abstract
Anaplastic meningioma is classified as a World Health Organization (WHO) grade III tumor and shows a strong tendency to recur. Although the incidence of anaplastic meningioma is low, the high rate of recurrence and death still makes treatment a challenge. A proteomics analysis was performed to investigate the differentially expressed proteins between anaplastic meningiomas and fibrous meningiomas by micro-LC-MS/MS. The key metabolic enzyme nicotinamide phosphoribosyltransferase (NAMPT) showed upregulated expression in anaplastic meningiomas. However, targeting NAMPT to treat anaplastic meningiomas has not been reported. In vitro, NAMPT inhibitor -FK866 reduced the viability of anaplastic meningiomas by inducing cell cycle arrest at the G2/M phase. Intriguingly, the NAMPT inhibitor -FK866 decreased the protein expression of immune checkpoints PD-L1 and B7-H3 by down-regulating the STAT1 and p-STAT1 expression in vitro. Furthermore, FK866 suppressed the growth of anaplastic meningiomas in an in vivo xenograft model. The expression of Ki-67 and immune checkpoint proteins (PD-L1 and B7-H3) showed significant differences between the group treated with FK866 and the control group treated with DMSO. In conclusion, the expression of NAMPT, which plays a crucial role in energy metabolism, was upregulated in anaplastic meningiomas. The NAMPT inhibitor -FK866 significantly suppressed the growth of anaplastic meningiomas in vitro and in vivo. More strikingly, FK866 potently inhibited immune checkpoint protein (PD-L1 and B7-H3) expression by regulating STAT1 in vitro and in vivo. Our results demonstrated that NAMPT inhibitors could potentially be an effective treatment method for patients suffering from anaplastic meningiomas.
Collapse
Affiliation(s)
- Yuxuan Deng
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Boyi Hu
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Yazhou Miao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jing Wang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Shaodong Zhang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Hong Wan
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Zhen Wu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yifan Lv
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jie Feng
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Nan Ji
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Deric Park
- Department of Neurology, University of Chicago Medical Center, Chicago, IL, United States
| | - Shuyu Hao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
21
|
Updated Functional Roles of NAMPT in Carcinogenesis and Therapeutic Niches. Cancers (Basel) 2022; 14:cancers14092059. [PMID: 35565188 PMCID: PMC9103253 DOI: 10.3390/cancers14092059] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/22/2022] [Accepted: 04/18/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary The advantages and applications of using the non-invasive way to detect serum biomarkers for assessing cancer diagnosis and prognosis have been explored. Nicotinamide phosphoribosyltransferase (NAMPT), also designated as pre-B-cell colony-enhancing factor (PBEF) or visfatin, is a secreted adipokine known to modulate tumor malignancies. Its significance in predicting cancer patient’s survival outcome further renders the implementation of NAMPT in clinical practice. In this review, recent discoveries of NAMPT in cancer studies were focused and integrated. We aim to provide updates for researchers who are proposing relevant objectives. Abstract Nicotinamide phosphoribosyltransferase (NAMPT) is notable for its regulatory roles in tumor development and progression. Emerging evidence regarding NAMPT somatic mutations in cancer patients, NAMPT expressional signatures in normal tissues and cancers, and the prognostic significance of NAMPT in many cancer types has attracted attention, and NAMPT is considered a potential biomarker of cancer. Recent discoveries have demonstrated the indirect association and direct biological functions of NAMPT in modulating cancer metastasis, proliferation, angiogenesis, cancer stemness, and chemoresistance to anticancer drugs. These findings warrant further investigation of the underlying mechanisms to provide knowledge for developing novel cancer therapeutics. In this review article, we explore recent research developments involving the oncogenic activities of NAMPT by summarizing current knowledge regarding NAMPT somatic mutations, clinical trials, transcriptome data, and clinical information and discoveries related to the NAMPT-induced signaling pathway in modulating hallmarks of cancer. Furthermore, the comprehensive representation of NAMPT RNA expression in a pancancer panel as well as in specific normal cell types at single-cell level are demonstrated. The results suggest potential sites and cell types that could facilitate NAMPT-related tumorigenesis. With this review, we aim to shed light on the regulatory roles of NAMPT in tumor development and progression, and provide information to guide future research directions in this field.
Collapse
|
22
|
Understanding Molecular Mechanisms of Phenotype Switching and Crosstalk with TME to Reveal New Vulnerabilities of Melanoma. Cells 2022; 11:cells11071157. [PMID: 35406721 PMCID: PMC8997563 DOI: 10.3390/cells11071157] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/20/2022] [Accepted: 03/21/2022] [Indexed: 12/15/2022] Open
Abstract
Melanoma cells are notorious for their high plasticity and ability to switch back and forth between various melanoma cell states, enabling the adaptation to sub-optimal conditions and therapeutics. This phenotypic plasticity, which has gained more attention in cancer research, is proposed as a new paradigm for melanoma progression. In this review, we provide a detailed and deep comprehensive recapitulation of the complex spectrum of phenotype switching in melanoma, the key regulator factors, the various and new melanoma states, and corresponding signatures. We also present an extensive description of the role of epigenetic modifications (chromatin remodeling, methylation, and activities of long non-coding RNAs/miRNAs) and metabolic rewiring in the dynamic switch. Furthermore, we elucidate the main role of the crosstalk between the tumor microenvironment (TME) and oxidative stress in the regulation of the phenotype switching. Finally, we discuss in detail several rational therapeutic approaches, such as exploiting phenotype-specific and metabolic vulnerabilities and targeting components and signals of the TME, to improve the response of melanoma patients to treatments.
Collapse
|
23
|
Audrito V, Moiso E, Ugolini F, Messana VG, Brandimarte L, Manfredonia I, Bianchi S, De Logu F, Nassini R, Szumera-Ciećkiewicz A, Taverna D, Massi D, Deaglio S. Tumors carrying BRAF-mutations over-express NAMPT that is genetically amplified and possesses oncogenic properties. J Transl Med 2022; 20:118. [PMID: 35272691 PMCID: PMC8908704 DOI: 10.1186/s12967-022-03315-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 02/19/2022] [Indexed: 01/01/2023] Open
Abstract
Background Nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme in nicotinamide adenine dinucleotide (NAD) biosynthesis, is up-regulated in several cancers, including metastatic melanoma (MM). The BRAF oncogene is mutated in different cancer types, among which MM and thyroid carcinoma (THCA) are prominent. Drugs targeting mutant BRAF are effective, especially in MM patients, even though resistance rapidly develops. Previous data have linked NAMPT over-expression to the acquisition of BRAF resistance, paving the way for therapeutic strategies targeting the two pathways. Methods Exploiting the TCGA database and a collection of MM and THCA tissue microarrays we studied the association between BRAF mutations and NAMPT expression. BRAF wild-type (wt) cell lines were genetically engineered to over-express the BRAF V600E construct to demonstrate a direct relationship between over-activation of the BRAF pathway and NAMPT expression. Responses of different cell line models to NAMPT (i)nhibitors were studied using dose–response proliferation assays. Analysis of NAMPT copy number variation was performed in the TCGA dataset. Lastly, growth and colony forming assays were used to study the tumorigenic functions of NAMPT itself. Results The first finding of this work is that tumor samples carrying BRAF-mutations over-express NAMPT, as demonstrated by analyzing the TCGA dataset, and MM and THC tissue microarrays. Importantly, BRAF wt MM and THCA cell lines modified to over-express the BRAF V600E construct up-regulated NAMPT, confirming a transcriptional regulation of NAMPT following BRAF oncogenic signaling activation. Treatment of BRAF-mutated cell lines with two different NAMPTi was followed by significant reduction of tumor growth, indicating NAMPT addiction in these cells. Lastly, we found that several tumors over-expressing the enzyme, display NAMPT gene amplification. Over-expression of NAMPT in BRAF wt MM cell line and in fibroblasts resulted in increased growth capacity, arguing in favor of oncogenic properties of NAMPT. Conclusions Overall, the association between BRAF mutations and NAMPT expression identifies a subset of tumors more sensitive to NAMPT inhibition opening the way for novel combination therapies including NAMPTi with BRAFi/MEKi, to postpone and/or overcome drug resistance. Lastly, the over-expression of NAMPT in several tumors could be a key and broad event in tumorigenesis, substantiated by the finding of NAMPT gene amplification. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03315-9.
Collapse
Affiliation(s)
- Valentina Audrito
- Laboratory of Functional Genomics, Department of Medical Sciences, University of Turin, Via Nizza, 52, 10126, Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Enrico Moiso
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.,Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Filippo Ugolini
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Vincenzo Gianluca Messana
- Laboratory of Functional Genomics, Department of Medical Sciences, University of Turin, Via Nizza, 52, 10126, Torino, Italy
| | - Lorenzo Brandimarte
- Laboratory of Functional Genomics, Department of Medical Sciences, University of Turin, Via Nizza, 52, 10126, Torino, Italy
| | - Ilaria Manfredonia
- Laboratory of Functional Genomics, Department of Medical Sciences, University of Turin, Via Nizza, 52, 10126, Torino, Italy
| | - Simonetta Bianchi
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Francesco De Logu
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Romina Nassini
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Anna Szumera-Ciećkiewicz
- Department of Pathology and Laboratory Medicine, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland.,Diagnostic Hematology Department, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Daniela Taverna
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Daniela Massi
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Silvia Deaglio
- Laboratory of Functional Genomics, Department of Medical Sciences, University of Turin, Via Nizza, 52, 10126, Torino, Italy.
| |
Collapse
|
24
|
Liu J, Tao X, Zhu Y, Li C, Ruan K, Diaz-Perez Z, Rai P, Wang H, Zhai RG. NMNAT promotes glioma growth through regulating post-translational modifications of P53 to inhibit apoptosis. eLife 2021; 10:70046. [PMID: 34919052 PMCID: PMC8683086 DOI: 10.7554/elife.70046] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 11/10/2021] [Indexed: 12/31/2022] Open
Abstract
Gliomas are highly malignant brain tumors with poor prognosis and short survival. NAD+ has been shown to impact multiple processes that are dysregulated in cancer; however, anti-cancer therapies targeting NAD+ synthesis have had limited success due to insufficient mechanistic understanding. Here, we adapted a Drosophila glial neoplasia model and discovered the genetic requirement for NAD+ synthase nicotinamide mononucleotide adenylyltransferase (NMNAT) in glioma progression in vivo and in human glioma cells. Overexpressing enzymatically active NMNAT significantly promotes glial neoplasia growth and reduces animal viability. Mechanistic analysis suggests that NMNAT interferes with DNA damage-p53-caspase-3 apoptosis signaling pathway by enhancing NAD+-dependent posttranslational modifications (PTMs) poly(ADP-ribosyl)ation (PARylation) and deacetylation of p53. Since PARylation and deacetylation reduce p53 pro-apoptotic activity, modulating p53 PTMs could be a key mechanism by which NMNAT promotes glioma growth. Our findings reveal a novel tumorigenic mechanism involving protein complex formation of p53 with NAD+ synthetic enzyme NMNAT and NAD+-dependent PTM enzymes that regulates glioma growth.
Collapse
Affiliation(s)
- Jiaqi Liu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai UniversityShandongChina
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of MedicineMiamiUnited States
| | - Xianzun Tao
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of MedicineMiamiUnited States
| | - Yi Zhu
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of MedicineMiamiUnited States
| | - Chong Li
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of MedicineMiamiUnited States
| | - Kai Ruan
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of MedicineMiamiUnited States
| | - Zoraida Diaz-Perez
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of MedicineMiamiUnited States
| | - Priyamvada Rai
- Department of Radiation Oncology, University of Miami Miller School of MedicineMiamiUnited States
- Sylvester Comprehensive Cancer CenterMiamiUnited States
| | - Hongbo Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai UniversityShandongChina
| | - R Grace Zhai
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of MedicineMiamiUnited States
- Sylvester Comprehensive Cancer CenterMiamiUnited States
| |
Collapse
|
25
|
Abstract
There has been considerable research in recent years on dietary factors and/or nutritional elements that might impact melanoma risk. A wide variety of dietary compounds have been studied, but only a selected group will be discussed in this review. Many have promising in vitro evidence supporting their potential, and some have been associated with decreased melanoma risk in epidemiologic studies; however, data from randomized controlled trials in humans are lacking. Future studies may be able to clarify the potential role of dietary components in melanoma risk reduction.
Collapse
|
26
|
Berthier-Vergnes O, Barbollat-Boutrand L, Pommier RM, de la Fouchardière A, Combemale P, Grimont M, Lopez-Ramirez N, Caramel J, Dalle S, Perrot JL, Gaudy-Marqueste C, Macagno N, Mansard S, Bouquet F, Masse I. Tetraspanin8 expression predicts an increased metastatic risk and is associated with cancer-related death in human cutaneous melanoma. Mol Cancer 2021; 20:127. [PMID: 34600553 PMCID: PMC8487126 DOI: 10.1186/s12943-021-01429-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 09/19/2021] [Indexed: 11/30/2022] Open
Affiliation(s)
- Odile Berthier-Vergnes
- CNRS, UMR5534, Centre de Génétique et de Physiologie Moléculaires et Cellulaires, F-69622, Villeurbanne, France
| | - Laetitia Barbollat-Boutrand
- Centre de Recherche en Cancérologie de Lyon, UMR Inserm 1052 CNRS 5286 Centre Leon Berard, Batiment Cheney D, 2eme etage, 28 rue Laennec, 69373 Cedex 08, Lyon, France
| | - Roxane M Pommier
- Centre de Recherche en Cancérologie de Lyon, UMR Inserm 1052 CNRS 5286 Centre Leon Berard, Batiment Cheney D, 2eme etage, 28 rue Laennec, 69373 Cedex 08, Lyon, France
| | - Arnaud de la Fouchardière
- Centre de Recherche en Cancérologie de Lyon, UMR Inserm 1052 CNRS 5286 Centre Leon Berard, Batiment Cheney D, 2eme etage, 28 rue Laennec, 69373 Cedex 08, Lyon, France
- Département de Biopathologie, Centre Leon Bérard, Lyon, France
| | | | - Maxime Grimont
- Centre de Recherche en Cancérologie de Lyon, UMR Inserm 1052 CNRS 5286 Centre Leon Berard, Batiment Cheney D, 2eme etage, 28 rue Laennec, 69373 Cedex 08, Lyon, France
| | - Noémie Lopez-Ramirez
- Centre de Recherche en Cancérologie de Lyon, UMR Inserm 1052 CNRS 5286 Centre Leon Berard, Batiment Cheney D, 2eme etage, 28 rue Laennec, 69373 Cedex 08, Lyon, France
| | - Julie Caramel
- Centre de Recherche en Cancérologie de Lyon, UMR Inserm 1052 CNRS 5286 Centre Leon Berard, Batiment Cheney D, 2eme etage, 28 rue Laennec, 69373 Cedex 08, Lyon, France
| | - Stéphane Dalle
- Centre de Recherche en Cancérologie de Lyon, UMR Inserm 1052 CNRS 5286 Centre Leon Berard, Batiment Cheney D, 2eme etage, 28 rue Laennec, 69373 Cedex 08, Lyon, France
| | - Jean-Luc Perrot
- Department of Dermatology, University Hospital of St-Etienne, Saint-Etienne, France
| | - Caroline Gaudy-Marqueste
- Dermatology and Skin Cancer Department, Aix-Marseille University, 264, rue Saint-Pierre, 13385, Marseille, France
| | - Nicolas Macagno
- Dermatology and Skin Cancer Department, Aix-Marseille University, 264, rue Saint-Pierre, 13385, Marseille, France
| | - Sandrine Mansard
- Department of Dermatology, University Hospital Estaing, Clermont Ferrand, France
| | | | - Ingrid Masse
- CNRS, UMR5534, Centre de Génétique et de Physiologie Moléculaires et Cellulaires, F-69622, Villeurbanne, France.
- Centre de Recherche en Cancérologie de Lyon, UMR Inserm 1052 CNRS 5286 Centre Leon Berard, Batiment Cheney D, 2eme etage, 28 rue Laennec, 69373 Cedex 08, Lyon, France.
| |
Collapse
|
27
|
Pisibon C, Ouertani A, Bertolotto C, Ballotti R, Cheli Y. Immune Checkpoints in Cancers: From Signaling to the Clinic. Cancers (Basel) 2021; 13:cancers13184573. [PMID: 34572799 PMCID: PMC8468441 DOI: 10.3390/cancers13184573] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 12/19/2022] Open
Abstract
The immune system is known to help fight cancers. Ten years ago, the first immune checkpoint inhibitor targeting CTLA4 was approved by the FDA to treat patients with metastatic melanoma. Since then, immune checkpoint therapies have revolutionized the field of oncology and the treatment of cancer patients. Numerous immune checkpoint inhibitors have been developed and tested, alone or in combination with other treatments, in melanoma and other cancers, with overall clear benefits to patient outcomes. However, many patients fail to respond or develop resistance to these treatments. It is therefore essential to decipher the mechanisms of action of immune checkpoints and to understand how immune cells are affected by signaling to be able to understand and overcome resistance. In this review, we discuss the signaling and effects of each immune checkpoint on different immune cells and their biological and clinical relevance. Restoring the functionality of T cells and their coordination with other immune cells is necessary to overcome resistance and help design new clinical immunotherapy strategies. In this respect, NK cells have recently been implicated in the resistance to anti-PD1 evoked by a protein secreted by melanoma, ITGBL1. The complexity of this network will have to be considered to improve the efficiency of future immunotherapies and may lead to the discovery of new immune checkpoints.
Collapse
Affiliation(s)
- Céline Pisibon
- Université Côte d’Azur, 06103 Nice, France; (C.P.); (A.O.); (C.B.); (R.B.)
- INSERM, Centre Méditerranéen de Médecine Moléculaire, Biology and Pathologies of Melanocytes, Team1, 06200 Nice, France
| | - Amira Ouertani
- Université Côte d’Azur, 06103 Nice, France; (C.P.); (A.O.); (C.B.); (R.B.)
- INSERM, Centre Méditerranéen de Médecine Moléculaire, Biology and Pathologies of Melanocytes, Team1, 06200 Nice, France
| | - Corine Bertolotto
- Université Côte d’Azur, 06103 Nice, France; (C.P.); (A.O.); (C.B.); (R.B.)
- INSERM, Centre Méditerranéen de Médecine Moléculaire, Biology and Pathologies of Melanocytes, Team1, 06200 Nice, France
| | - Robert Ballotti
- Université Côte d’Azur, 06103 Nice, France; (C.P.); (A.O.); (C.B.); (R.B.)
- INSERM, Centre Méditerranéen de Médecine Moléculaire, Biology and Pathologies of Melanocytes, Team1, 06200 Nice, France
| | - Yann Cheli
- Université Côte d’Azur, 06103 Nice, France; (C.P.); (A.O.); (C.B.); (R.B.)
- INSERM, Centre Méditerranéen de Médecine Moléculaire, Biology and Pathologies of Melanocytes, Team1, 06200 Nice, France
- Correspondence:
| |
Collapse
|
28
|
Houlier A, Pissaloux D, Tirode F, Lopez Ramirez N, Plaschka M, Caramel J, Masse I, de la Fouchardiere A. RASGRF2 gene fusions identified in a variety of melanocytic lesions with distinct morphological features. Pigment Cell Melanoma Res 2021; 34:1074-1083. [PMID: 34310073 DOI: 10.1111/pcmr.13004] [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: 04/12/2021] [Revised: 07/11/2021] [Accepted: 07/16/2021] [Indexed: 11/29/2022]
Abstract
The WHO classification identifies nine classes of melanocytic proliferations according to location, UV exposure, histological, and genetic features. Only a minority of lesions remain unclassified. We describe five cases that harbored either an ERBIN-RASGRF2 or an ATP2B4-RASGRF2 in-frame fusion transcript. These lesions were collected from different studies, unified only by the lack of identifiable known mutations, with a highly variable phenotype. One case was a large abdominal congenital nevus, three were slowly growing pigmented nodules, and the last was an ulcerated nodule arising on the site of a preexisting small nevus, known since childhood. The latter was diagnosed as a 4 mm thick melanoma with loss of BAP1 expression. The four other cases were compound, melanocytic proliferations with an unusual deep pattern of small dense nests of bland melanocytes encased in a fibrous background. The RASGRF2 fusion was confirmed by a break-apart FISH technique. Array CGH performed in three cases found non-recurrent secondary copy number alterations. Follow-up was uneventful. In silico analysis identified a single RASGRF2 fusion in the TCGA pan-cancer database, whereas RASGRF2 variants were stochastically distributed in all cancer subtypes.
Collapse
Affiliation(s)
- Aurélie Houlier
- University of Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5286, INSERM U1052, Cancer Research Centre of Lyon, Lyon, France.,Department of Biopathology, Centre Léon Bérard, Lyon, France
| | - Daniel Pissaloux
- University of Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5286, INSERM U1052, Cancer Research Centre of Lyon, Lyon, France.,Department of Biopathology, Centre Léon Bérard, Lyon, France
| | - Franck Tirode
- University of Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5286, INSERM U1052, Cancer Research Centre of Lyon, Lyon, France
| | - Noémie Lopez Ramirez
- University of Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5286, INSERM U1052, Cancer Research Centre of Lyon, Lyon, France
| | - Maud Plaschka
- University of Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5286, INSERM U1052, Cancer Research Centre of Lyon, Lyon, France
| | - Julie Caramel
- University of Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5286, INSERM U1052, Cancer Research Centre of Lyon, Lyon, France
| | - Ingrid Masse
- University of Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5286, INSERM U1052, Cancer Research Centre of Lyon, Lyon, France
| | - Arnaud de la Fouchardiere
- University of Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5286, INSERM U1052, Cancer Research Centre of Lyon, Lyon, France.,Department of Biopathology, Centre Léon Bérard, Lyon, France
| |
Collapse
|
29
|
Metabolic Interplay between the Immune System and Melanoma Cells: Therapeutic Implications. Biomedicines 2021; 9:biomedicines9060607. [PMID: 34073463 PMCID: PMC8227307 DOI: 10.3390/biomedicines9060607] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/20/2021] [Accepted: 05/25/2021] [Indexed: 02/06/2023] Open
Abstract
Malignant melanoma represents the most fatal skin cancer due to its aggressive biological behavior and high metastatic potential. Treatment strategies for advanced disease have dramatically changed over the last years due to the introduction of BRAF/MEK inhibitors and immunotherapy. However, many patients either display primary (i.e., innate) or eventually develop secondary (i.e., acquired) resistance to systemic treatments. Treatment resistance depends on multiple mechanisms driven by a set of rewiring processes, which involve cancer metabolism, epigenetic, gene expression, and interactions within the tumor microenvironment. Prognostic and predictive biomarkers are needed to guide patients’ selection and treatment decisions. Indeed, there are no recognized clinical or biological characteristics that identify which patients will benefit more from available treatments, but several biomarkers have been studied with promising preliminary results. In this review, we will summarize novel tumor metabolic pathways and tumor-host metabolic crosstalk mechanisms leading to melanoma progression and drug resistance, with an overview on their translational potential as novel therapeutic targets.
Collapse
|
30
|
Zhang F, Tang X, Fan S, Liu X, Sun J, Ju C, Liang Y, Liu R, Zhou R, Yu B, Zhang C, Zhang Z, Kang T, Huang G, Lv XB. Targeting the p300/NONO axis sensitizes melanoma cells to BRAF inhibitors. Oncogene 2021; 40:4137-4150. [PMID: 34017080 DOI: 10.1038/s41388-021-01834-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 04/25/2021] [Accepted: 05/07/2021] [Indexed: 01/01/2023]
Abstract
BRAF inhibitors (BRAFi) that target BRAF V600E kinase, a driver mutation found in 50% of melanomas, show a significant antitumor response, but the common emergence of acquired resistance remains a challenge. Abnormal expression of RAF isoforms CRAF and ARAF reactivates pERK1/2, which plays crucial roles in the acquisition of resistance of melanoma cells. However, the mechanisms of dysregulation of RAF isoforms in resistant melanoma cells remain unknown. Here, we identified NONO interacted with and stabilized both CRAF and ARAF in melanoma cells, and that NONO was acetylated at 198K by p300 acetyltransferase, which stabilized NONO via antagonizing its ubiquitination/degradation mediated by RNF8. The upregulation of both p300 and NONO promoted the rebound of pERK1/2 and the subsequent resistance of melanoma cells to BRAFi, and the activation of ERK1/2 in turn induced p300 to form a positive feedback loop in resistant melanoma cells. There was a positive correlation between p300 and NONO in resistant melanoma cells and clinical samples, and p300 inhibitor C646 overcame the resistance of resistant melanoma cells to BRAF inhibitors in vitro and in vivo. Our findings reveal that targeting the positive feedback loop of p300-NONO-CRAF/ARAF-pERK1/2 may be excellent strategies to overcome the resistance of BRAF inhibitors for melanoma patients.
Collapse
Affiliation(s)
- Feifei Zhang
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The Third Affiliated Hospital of Nanchang University, Nanchang, PR China
| | - Xiaofeng Tang
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The Third Affiliated Hospital of Nanchang University, Nanchang, PR China
| | - Song Fan
- Department of Oral and Maxillofacial Surgery, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, PR China
| | - Xia Liu
- Department of Medical Oncology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, PR China
| | - Jun Sun
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The Third Affiliated Hospital of Nanchang University, Nanchang, PR China.,College of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, PR China
| | - Cheng Ju
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The Third Affiliated Hospital of Nanchang University, Nanchang, PR China.,Department of Orthopedics, The Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, PR China
| | - Yiping Liang
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The Third Affiliated Hospital of Nanchang University, Nanchang, PR China
| | - Renfeng Liu
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The Third Affiliated Hospital of Nanchang University, Nanchang, PR China.,Department of Orthopedics, The Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, PR China
| | - Ruihao Zhou
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The Third Affiliated Hospital of Nanchang University, Nanchang, PR China
| | - Bo Yu
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The Third Affiliated Hospital of Nanchang University, Nanchang, PR China.,Department of Orthopedics, The Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, PR China
| | - Changhua Zhang
- College of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, PR China
| | - Zhiping Zhang
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The Third Affiliated Hospital of Nanchang University, Nanchang, PR China.,Department of Orthopedics, The Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, PR China
| | - Tiebang Kang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, PR China
| | - Guofu Huang
- Department of Ophthalmology, The Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, PR China.
| | - Xiao-Bin Lv
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The Third Affiliated Hospital of Nanchang University, Nanchang, PR China.
| |
Collapse
|
31
|
Huffaker TB, Ekiz HA, Barba C, Lee SH, Runtsch MC, Nelson MC, Bauer KM, Tang WW, Mosbruger TL, Cox JE, Round JL, Voth WP, O'Connell RM. A Stat1 bound enhancer promotes Nampt expression and function within tumor associated macrophages. Nat Commun 2021; 12:2620. [PMID: 33976173 PMCID: PMC8113251 DOI: 10.1038/s41467-021-22923-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 03/30/2021] [Indexed: 02/03/2023] Open
Abstract
Tumor associated macrophage responses are regulated by distinct metabolic states that affect their function. However, the ability of specific signals in the local tumor microenvironment to program macrophage metabolism remains under investigation. Here, we identify NAMPT, the rate limiting enzyme in NAD salvage synthesis, as a target of STAT1 during cellular activation by interferon gamma, an important driver of macrophage polarization and antitumor responses. We demonstrate that STAT1 occupies a conserved element within the first intron of Nampt, termed Nampt-Regulatory Element-1 (NRE1). Through disruption of NRE1 or pharmacological inhibition, a subset of M1 genes is sensitive to NAMPT activity through its impact on glycolytic processes. scRNAseq is used to profile in vivo responses by NRE1-deficient, tumor-associated leukocytes in melanoma tumors through the creation of a unique mouse strain. Reduced Nampt and inflammatory gene expression are present in specific myeloid and APC populations; moreover, targeted ablation of NRE1 in macrophage lineages results in greater tumor burden. Finally, elevated NAMPT expression correlates with IFNγ responses and melanoma patient survival. This study identifies IFN and STAT1-inducible Nampt as an important factor that shapes the metabolic program and function of tumor associated macrophages.
Collapse
Affiliation(s)
- Thomas B Huffaker
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - H Atakan Ekiz
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Cindy Barba
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Soh-Hyun Lee
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Marah C Runtsch
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Morgan C Nelson
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Kaylyn M Bauer
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - William W Tang
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | | | - James E Cox
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
- Metabolomics Core Research Facility, University of Utah, Salt Lake City, UT, USA
| | - June L Round
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Warren P Voth
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, USA.
| | - Ryan M O'Connell
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, USA.
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
32
|
Olszańska J, Pietraszek-Gremplewicz K, Nowak D. Melanoma Progression under Obesity: Focus on Adipokines. Cancers (Basel) 2021; 13:cancers13092281. [PMID: 34068679 PMCID: PMC8126042 DOI: 10.3390/cancers13092281] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/01/2021] [Accepted: 05/05/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Obesity is a rapidly growing public health problem and the reason for numerous diseases in the human body, including cancer. This article reviews the current knowledge of the effect of molecules secreted by adipose tissue-adipokines on melanoma progression. We also discuss the role of these factors as markers of incidence, metastasis, and melanoma patient survival. Understanding the functions of adipokines will lead to knowledge of whether and how obesity promotes melanoma growth. Abstract Obesity is a growing problem in the world and is one of the risk factors of various cancers. Among these cancers is melanoma, which accounts for the majority of skin tumor deaths. Current studies are looking for a correlation between obesity and melanoma. They suspect that a potential cause of its development is connected to the biology of adipokines, active molecules secreted by adipose tissue. Under physiological conditions, adipokines control many processes, including lipid and glucose homeostasis, insulin sensitivity, angiogenesis, and inflammations. However, when there is an increased amount of fat in the body, their secretion is dysregulated. This article reviews the current knowledge of the effect of adipokines on melanoma growth. This work focuses on the molecular pathways by which adipose tissue secreted molecules modify the angiogenesis, migration, invasion, proliferation, and death of melanoma cells. We also discuss the role of these factors as markers of incidence, metastasis, and melanoma patient survival. Understanding the functions of adipokines will lead to knowledge of whether and how obesity promotes melanoma growth. Further studies may contribute to the innovations of therapies and the use of adipokines as predictive and/or prognostic biomarkers.
Collapse
|
33
|
Patton EE, Mueller KL, Adams DJ, Anandasabapathy N, Aplin AE, Bertolotto C, Bosenberg M, Ceol CJ, Burd CE, Chi P, Herlyn M, Holmen SL, Karreth FA, Kaufman CK, Khan S, Kobold S, Leucci E, Levy C, Lombard DB, Lund AW, Marie KL, Marine JC, Marais R, McMahon M, Robles-Espinoza CD, Ronai ZA, Samuels Y, Soengas MS, Villanueva J, Weeraratna AT, White RM, Yeh I, Zhu J, Zon LI, Hurlbert MS, Merlino G. Melanoma models for the next generation of therapies. Cancer Cell 2021; 39:610-631. [PMID: 33545064 PMCID: PMC8378471 DOI: 10.1016/j.ccell.2021.01.011] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 12/12/2022]
Abstract
There is a lack of appropriate melanoma models that can be used to evaluate the efficacy of novel therapeutic modalities. Here, we discuss the current state of the art of melanoma models including genetically engineered mouse, patient-derived xenograft, zebrafish, and ex vivo and in vitro models. We also identify five major challenges that can be addressed using such models, including metastasis and tumor dormancy, drug resistance, the melanoma immune response, and the impact of aging and environmental exposures on melanoma progression and drug resistance. Additionally, we discuss the opportunity for building models for rare subtypes of melanomas, which represent an unmet critical need. Finally, we identify key recommendations for melanoma models that may improve accuracy of preclinical testing and predict efficacy in clinical trials, to help usher in the next generation of melanoma therapies.
Collapse
Affiliation(s)
- E Elizabeth Patton
- MRC Human Genetics Unit and Cancer Research UK Edinburgh Centre, MRC Institute of Genetics & Molecular Medicine, The University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK.
| | - Kristen L Mueller
- Melanoma Research Alliance, 730 15th Street NW, Washington, DC 20005, USA.
| | - David J Adams
- Experimental Cancer Genetics, Wellcome Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | - Niroshana Anandasabapathy
- Department of Dermatology, Meyer Cancer Center, Program in Immunology and Microbial Pathogenesis, Weill Cornell Medicine, New York, NY 10026, USA
| | - Andrew E Aplin
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Corine Bertolotto
- Université Côte d'Azur, Nice, France; INSERM, Biology and Pathologies of Melanocytes, Team 1, Equipe Labellisée Ligue 2020, Centre Méditerranéen de Médecine Moléculaire, Nice, France
| | - Marcus Bosenberg
- Departments of Dermatology, Pathology, and Immunobiology, Yale University, New Haven, CT, USA
| | - Craig J Ceol
- Program in Molecular Medicine and Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Christin E Burd
- Departments of Molecular Genetics, Cancer Biology, and Genetics, The Ohio State University, Biomedical Research Tower, Room 918, 460 W. 12th Avenue, Columbus, OH 43210, USA
| | - Ping Chi
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | | | - Sheri L Holmen
- Department of Surgery, University of Utah Health Sciences Center, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Florian A Karreth
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Charles K Kaufman
- Washington University School of Medicine, Department of Medicine, Division of Oncology, Department of Developmental Biology, McDonnell Science Building, 4518 McKinley Avenue, St. Louis, MO 63110, USA
| | - Shaheen Khan
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Sebastian Kobold
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU, Munich, Germany; Member of the German Center for Lung Research (DZL), German Center for Translational Cancer Research (DKTK), partner site Munich, Munich, Germany
| | - Eleonora Leucci
- Laboratory for RNA Cancer Biology, Department of Oncology, LKI, KU Leuven, 3000 Leuven, Belgium; Trace, Department of Oncology, LKI, KU Leuven, 3000 Leuven, Belgium
| | - Carmit Levy
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - David B Lombard
- Department of Pathology, Institute of Gerontology, and Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Amanda W Lund
- Ronald O. Perelman Department of Dermatology and Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Kerrie L Marie
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jean-Christophe Marine
- Laboratory for Molecular Cancer Biology, Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory for Molecular Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Richard Marais
- CRUK Manchester Institute, The University of Manchester, Alderley Park, Macclesfield SK10 4TG, UK
| | - Martin McMahon
- Department of Dermatology & Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Carla Daniela Robles-Espinoza
- Laboratorio Internacional de Investigación sobre el Genoma Humano, Universidad Nacional Autónoma de México, Campus Juriquilla, Boulevard Juriquilla 3001, Santiago de Querétaro 76230, Mexico; Wellcome Sanger Institute, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Ze'ev A Ronai
- Cancer Center, Sanford Burnham Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Yardena Samuels
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Maria S Soengas
- Spanish National Cancer Research Centre, 28029 Madrid, Spain
| | - Jessie Villanueva
- The Wistar Institute, Molecular and Cellular Oncogenesis Program, Philadelphia, PA, USA
| | - Ashani T Weeraratna
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, and Department of Oncology, Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Richard M White
- Department of Cancer Biology & Genetics and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Iwei Yeh
- Departments of Dermatology and Pathology, University of California, San Francisco, CA, USA
| | - Jiyue Zhu
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, USA
| | - Leonard I Zon
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Medical School, Harvard Stem Cell Institute, Stem Cell and Regenerative Biology Department, Harvard University, Boston, MA, USA
| | - Marc S Hurlbert
- Melanoma Research Alliance, 730 15th Street NW, Washington, DC 20005, USA
| | - Glenn Merlino
- Center for Cancer Research, NCI, NIH, 37 Convent Drive, Bethesda, MD 20892, USA.
| |
Collapse
|
34
|
Annie L, Gurusubramanian G, Roy VK. Inhibition of visfatin by FK866 mitigates pathogenesis of cystic ovary in letrozole-induced hyperandrogenised mice. Life Sci 2021; 276:119409. [PMID: 33781825 DOI: 10.1016/j.lfs.2021.119409] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/08/2021] [Accepted: 03/18/2021] [Indexed: 02/07/2023]
Abstract
Polycystic ovary syndrome is a common reproductive disorder in the female of reproductive age, which is characterized by hyperandrogenism, insulin resistance, cystic ovary and infertility. The level of pro-inflammatory adipokine, visfatin is elevated in PCOS conditions in human and animal. In this study, letrozole induced hyperandrogenised PCOS mice model have been used to unravel the effects of visfatin inhibition. The results showed that letrozole induced hyperandrogenisation significantly (p < 0.05) elevates ovarian visfatin concentration from 66.03 ± 1.77 to 112.08 ± 3.7 ng/ml, and visfatin expression to 2.5 fold (p < 0.05) compared to control. Visfatin inhibition in PCOS by FK866 has significantly (p < 0.05) suppressed the secretion of androgens, androstenedione (from 0.329 ± 0.07 to 0.097 ± 0.01 ng/ml) and testosterone levels (from 0.045 ± 0.003 to 0.014 ± 0.0009 ng/ml). Ovarian histology showed that visfatin inhibition suppressed cyst formation and promotes corpus luteum formation. Visfatin inhibition has suppressed apoptosis and increases the expression of BCL2 along with increase in the proliferation (GCNA expression elevated). Visfatin inhibition has increased ovarian glucose content (from 167.05 ± 8.5 to 210 ± 7 mg/dl), along with increase in ovarian GLUT8 expression. In vitro study has also supported the in vivo findings where FK866 treatment significantly (p < 0.05) suppressed testosterone (control-3.84 ± 0.44 ng/ml, 1 nM FK866-2.02 ± 0.048 ng/ml, 10 nM FK866-1.74 ± 0.20 ng/ml) and androstenedione (control-4.68 ± 0.91 ng/ml, 1 nM FK866-3.38 ± 0.27 ng/ml, 10 nM FK866-4.55 ± 0.83 ng/ml) production from PCOS ovary. In conclusion, this is first report, which showed that visfatin inhibition by FK866 in hyperandrogenised mice ameliorates pathogenesis of PCOS. Thus, it may be suggested that visfatin inhibition could have a therapeutic potential in PCOS management along with other intervention.
Collapse
Affiliation(s)
| | | | - Vikas Kumar Roy
- Department of Zoology, Mizoram University, Aizawl, Mizoram 796 004, India.
| |
Collapse
|
35
|
Liu J, Che X, You J, Zhang G, Zhao R, Fu J, Li H. Intracellular Nampt impairs esophageal squamous cell carcinoma neo-adjuvant chemotherapy response independent of eNampt. Am J Transl Res 2021; 13:1411-1421. [PMID: 33841666 PMCID: PMC8014354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/18/2021] [Indexed: 06/12/2023]
Abstract
Nampt consists of iNampt and eNampt, might contribute to modulating obesity-related malignancies and impairing response to chemotherapy in a range of cancers. This study explored the role of Nampt and adiposity in the progression and response to neo-adjuvant chemotherapy of esophageal squamous cell carcinoma (ESCC). Patients with ESCC were treated with 2 cycles of neo-adjuvant chemotherapy, then evaluated for surgery. Tumor regression grading (TRG) and prognosis of these patients were collected. Anthropometry was well utilized. Serum eNampt was determined by enzyme-linked immunosorbent assay, iNampt expression in tissues were assessed by PCR, western blot and immunohistochemistry. eNampt in sera elevated significantly in these over-weight or obese patients, and was positively associated with body mass index (BMI), waist circumference, visceral fat area (VFA), subcutaneous fat area (SFA) and total fat area (TFA) (P<0.05). iNampt expression in the mRNA and protein levels were up-regulated in ESCC compared to their adjacent non-tumor specimens (P<0.05). iNampt protein staining revealed mainly in the cytoplasm and nuclei, while it was not related to serum eNampt, BMI, waist circumference, VFA, SFA and TFA (P>0.05). Pre-treatment iNampt, BMI, SFA, TFA and age significantly correlated with neo-adjuvant chemotherapy response, and iNampt expression and age were independent predictors (P<0.05). Pre-treatment iNampt, ypT, ypN, ypTNM stage and TRG were associated with the survival of ESCCs, and ypN stage and TRG were independent prognostic factors (P<0.05). In conclusion, iNampt impaired ESCC response to neo-adjuvant chemotherapy independent of eNampt, targeting iNampt to increase ESCC response to neo-adjuvant chemotherapy would improve the prognosis of ESCCs.
Collapse
Affiliation(s)
- Jiahuang Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an, Shaanxi, China
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an, Shaanxi, China
| | - Xiangming Che
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an, Shaanxi, China
| | - Jiangtao You
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an, Shaanxi, China
| | - Guangjian Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an, Shaanxi, China
| | - Rui Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an, Shaanxi, China
| | - Junke Fu
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an, Shaanxi, China
| | - Haijun Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an, Shaanxi, China
| |
Collapse
|
36
|
Annie L, Gurusubramanian G, Kumar Roy V. Visfatin protein may be responsible for suppression of proliferation and apoptosis in the infantile mice ovary. Cytokine 2021; 140:155422. [PMID: 33476980 DOI: 10.1016/j.cyto.2021.155422] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 12/21/2020] [Accepted: 12/24/2020] [Indexed: 11/19/2022]
Abstract
Visfatin is an important adipokines, which are expressed in different tissues including ovary of mammals. The postnatal ovary in rodents undergoes dramatic changes of intra-ovarian factors in relation to proliferation and apoptosis. There are studies which showed that gonadal visfatin changes in postnatal life. However, role of visfatin in the early postnatal period i.e. infantile period has not been studied. Therefore, the present study was aimed to explore the role of visfatin in the early postnatal ovarian functions. Furthermore, to explore the role of visfatin, the endogenous visfatin was inhibited from PND14-PND21 by FK866 with dose of 1.5 mg/kg. Our results showed gain in body weight and ovarian weight after visfatin inhibition. The inhibition of visfatin increased the ovarian proliferation (increase in PCNA, GCNA expression and BrdU incorporation) and apoptosis (increase in BAX and active caspase3 expression). Moreover, visfatin inhibition decreased the expression of antiapoptotic/survival protein, BCL2 in the ovary. These findings suggest that visfatin in the infantile ovary may suppress the proliferation and apoptosis by up-regulating BCL2 expression. An interesting finding has been observed that circulating estrogen and progesterone remain unaffected, although visfatin inhibition up-regulated ER-β and down-regulated ER-α. It may also be suggested that visfatin could regulates proliferation and apoptosis via modulating estrogen signaling. In conclusion, visfatin inhibits the proliferation and apoptosis without modulating the ovarian steroid biosynthesis and visfatin mediated BCL2 expression could also be mechanism to preserve the good quality follicle in early postnatal period.
Collapse
Affiliation(s)
| | | | - Vikas Kumar Roy
- Department of Zoology, Mizoram University, Aizawl, Mizoram - 796 004, India.
| |
Collapse
|
37
|
FBXO32 links ubiquitination to epigenetic reprograming of melanoma cells. Cell Death Differ 2021; 28:1837-1848. [PMID: 33462405 PMCID: PMC8184796 DOI: 10.1038/s41418-020-00710-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 12/01/2020] [Accepted: 12/11/2020] [Indexed: 12/15/2022] Open
Abstract
Ubiquitination by serving as a major degradation signal of proteins, but also by controlling protein functioning and localization, plays critical roles in most key cellular processes. Here, we show that MITF, the master transcription factor in melanocytes, controls ubiquitination in melanoma cells. We identified FBXO32, a component of the SCF E3 ligase complex as a new MITF target gene. FBXO32 favors melanoma cell migration, proliferation, and tumor development in vivo. Transcriptomic analysis shows that FBXO32 knockdown induces a global change in melanoma gene expression profile. These include the inhibition of CDK6 in agreement with an inhibition of cell proliferation and invasion upon FBXO32 silencing. Furthermore, proteomic analysis identifies SMARC4, a component of the chromatin remodeling complexes BAF/PBAF, as a FBXO32 partner. FBXO32 and SMARCA4 co-localize at loci regulated by FBXO32, such as CDK6 suggesting that FBXO32 controls transcription through the regulation of chromatin remodeling complex activity. FBXO32 and SMARCA4 are the components of a molecular cascade, linking MITF to epigenetics, in melanoma cells.
Collapse
|
38
|
Lv H, Lv G, Chen C, Zong Q, Jiang G, Ye D, Cui X, He Y, Xiang W, Han Q, Tang L, Yang W, Wang H. NAD + Metabolism Maintains Inducible PD-L1 Expression to Drive Tumor Immune Evasion. Cell Metab 2021; 33:110-127.e5. [PMID: 33171124 DOI: 10.1016/j.cmet.2020.10.021] [Citation(s) in RCA: 158] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 08/04/2020] [Accepted: 10/21/2020] [Indexed: 12/16/2022]
Abstract
NAD+ metabolism is implicated in aging and cancer. However, its role in immune checkpoint regulation and immune evasion remains unclear. Here, we find nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme of the NAD+ biogenesis, drives interferon γ (IFNγ)-induced PD-L1 expression in multiple types of tumors and governs tumor immune evasion in a CD8+ T cell-dependent manner. Mechanistically, NAD+ metabolism maintains activity and expression of methylcytosine dioxygenase Tet1 via α-ketoglutarate (α-KG). IFNγ-activated Stat1 facilitates Tet1 binding to Irf1 to regulate Irf1 demethylation, leading to downstream PD-L1 expression on tumors. Importantly, high NAMPT-expressing tumors are more sensitive to anti-PD-L1 treatment and NAD+ augmentation enhances the efficacy of anti-PD-L1 antibody in immunotherapy-resistant tumors. Collectively, these data delineate an NAD+ metabolism-dependent epigenetic mechanism contributing to tumor immune evasion, and NAD+ replenishment combined with PD-(L)1 antibody provides a promising therapeutic strategy for immunotherapy-resistant tumors.
Collapse
Affiliation(s)
- Hongwei Lv
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China; National Center for Liver Cancer, Second Military Medical University, Shanghai 201805, China; Shanghai Key Laboratory of Hepato-biliary Tumor Biology, Shanghai 200438, China
| | - Guishuai Lv
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China; National Center for Liver Cancer, Second Military Medical University, Shanghai 201805, China; Ministry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer, Shanghai 200438, China
| | - Cian Chen
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China; National Center for Liver Cancer, Second Military Medical University, Shanghai 201805, China
| | - Qianni Zong
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China; National Center for Liver Cancer, Second Military Medical University, Shanghai 201805, China
| | - Guoqing Jiang
- Department of Hepatobiliary Surgery, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu 225000, China
| | - Dan Ye
- Molecular and Cell Biology Lab, Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xiuliang Cui
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China; National Center for Liver Cancer, Second Military Medical University, Shanghai 201805, China
| | - Yufei He
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China; National Center for Liver Cancer, Second Military Medical University, Shanghai 201805, China
| | - Wei Xiang
- Cancer Research Center, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Qin Han
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China; National Center for Liver Cancer, Second Military Medical University, Shanghai 201805, China
| | - Liang Tang
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China; National Center for Liver Cancer, Second Military Medical University, Shanghai 201805, China
| | - Wen Yang
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China; National Center for Liver Cancer, Second Military Medical University, Shanghai 201805, China.
| | - Hongyang Wang
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China; National Center for Liver Cancer, Second Military Medical University, Shanghai 201805, China; Cancer Research Center, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China; Fudan University Shanghai Cancer Center, Shanghai 200032, China.
| |
Collapse
|
39
|
Audrito V, Messana VG, Moiso E, Vitale N, Arruga F, Brandimarte L, Gaudino F, Pellegrino E, Vaisitti T, Riganti C, Piva R, Deaglio S. NAMPT Over-Expression Recapitulates the BRAF Inhibitor Resistant Phenotype Plasticity in Melanoma. Cancers (Basel) 2020; 12:cancers12123855. [PMID: 33419372 PMCID: PMC7766175 DOI: 10.3390/cancers12123855] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 12/15/2020] [Indexed: 12/31/2022] Open
Abstract
Simple Summary Malignant melanoma (MM) is the most fatal skin cancer due to its high metastatic potential. Treatment strategies are dramatically changing due to the introduction of BRAF/MEK inhibitors (i) and immunotherapy; however, multiple resistant mechanisms rapidly occur including metabolic rewiring. This study aimed to establish the driver role of the nicotinamide adenine dinucleotide (NAD)-biosynthetic enzyme nicotinamide phosphoribosyltransferase (NAMPT) in BRAFi resistance development. We defined that NAMPT over-expressing MM cells were strikingly similar to cells that acquired resistance to BRAFi in terms of growth, invasion, and phenotype plasticity. These findings confirmed NAMPT as a key factor in melanoma progression and in the onset of BRAFi resistance in melanoma patients, opening new therapeutic possibilities for this subset of patients. Abstract Serine–threonine protein kinase B-RAF (BRAF)-mutated metastatic melanoma (MM) is a highly aggressive type of skin cancer. Treatment of MM patients using BRAF/MEK inhibitors (BRAFi/MEKi) eventually leads to drug resistance, limiting any clinical benefit. Herein, we demonstrated that the nicotinamide adenine dinucleotide (NAD)-biosynthetic enzyme nicotinamide phosphoribosyltransferase (NAMPT) is a driving factor in BRAFi resistance development. Using stable and inducible NAMPT over-expression systems, we showed that forced NAMPT expression in MM BRAF-mutated cell lines led to increased energy production, MAPK activation, colony-formation capacity, and enhance tumorigenicity in vivo. Moreover, NAMPT over-expressing cells switched toward an invasive/mesenchymal phenotype, up-regulating expression of ZEB1 and TWIST, two transcription factors driving the epithelial to mesenchymal transition (EMT) process. Consistently, within the NAMPT-overexpressing cell line variants, we observed an increased percentage of a rare, drug-effluxing stem cell-like side population (SP) of cells, paralleled by up-regulation of ABCC1/MRP1 expression and CD133-positive cells. The direct correlation between NAMPT expression and gene set enrichments involving metastasis, invasiveness and mesenchymal/stemness properties were verified also in melanoma patients by analyzing The Cancer Genome Atlas (TCGA) datasets. On the other hand, CRISPR/Cas9 full knock-out NAMPT BRAFi-resistant MM cells are not viable, while inducible partial silencing drastically reduces tumor growth and aggressiveness. Overall, this work revealed that NAMPT over-expression is both necessary and sufficient to recapitulate the BRAFi-resistant phenotype plasticity.
Collapse
Affiliation(s)
- Valentina Audrito
- Cancer Immunogenetics Lab, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (V.G.M.); (N.V.); (F.A.); (L.B.); (F.G.); (T.V.)
- Correspondence: (V.A.); (S.D.); Tel.: +39-0116709535-37 (V.A. & S.D.)
| | - Vincenzo Gianluca Messana
- Cancer Immunogenetics Lab, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (V.G.M.); (N.V.); (F.A.); (L.B.); (F.G.); (T.V.)
| | - Enrico Moiso
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA;
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Nicoletta Vitale
- Cancer Immunogenetics Lab, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (V.G.M.); (N.V.); (F.A.); (L.B.); (F.G.); (T.V.)
| | - Francesca Arruga
- Cancer Immunogenetics Lab, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (V.G.M.); (N.V.); (F.A.); (L.B.); (F.G.); (T.V.)
| | - Lorenzo Brandimarte
- Cancer Immunogenetics Lab, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (V.G.M.); (N.V.); (F.A.); (L.B.); (F.G.); (T.V.)
| | - Federica Gaudino
- Cancer Immunogenetics Lab, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (V.G.M.); (N.V.); (F.A.); (L.B.); (F.G.); (T.V.)
| | - Elisa Pellegrino
- Department of Molecular Biotechnologies and Health Sciences, University of Turin, 10126 Turin, Italy; (E.P.); (R.P.)
| | - Tiziana Vaisitti
- Cancer Immunogenetics Lab, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (V.G.M.); (N.V.); (F.A.); (L.B.); (F.G.); (T.V.)
| | - Chiara Riganti
- Department of Oncology, University of Turin, 10126 Turin, Italy;
| | - Roberto Piva
- Department of Molecular Biotechnologies and Health Sciences, University of Turin, 10126 Turin, Italy; (E.P.); (R.P.)
| | - Silvia Deaglio
- Cancer Immunogenetics Lab, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (V.G.M.); (N.V.); (F.A.); (L.B.); (F.G.); (T.V.)
- Correspondence: (V.A.); (S.D.); Tel.: +39-0116709535-37 (V.A. & S.D.)
| |
Collapse
|
40
|
Ballotti R, Cheli Y, Bertolotto C. The complex relationship between MITF and the immune system: a Melanoma ImmunoTherapy (response) Factor? Mol Cancer 2020; 19:170. [PMID: 33276788 PMCID: PMC7718690 DOI: 10.1186/s12943-020-01290-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 11/29/2020] [Indexed: 12/18/2022] Open
Abstract
The clinical benefit of immune checkpoint inhibitory therapy (ICT) in advanced melanomas is limited by primary and acquired resistance. The molecular determinants of the resistance have been extensively studied, but these discoveries have not yet been translated into therapeutic benefits. As such, a paradigm shift in melanoma treatment, to surmount the therapeutic impasses linked to the resistance, is an important ongoing challenge.This review outlines the multifaceted interplay between microphthalmia-associated transcription factor (MITF), a major determinant of the biology of melanoma cells, and the immune system. In melanomas, MITF functions downstream oncogenic pathways and microenvironment stimuli that restrain the immune responses. We highlight how MITF, by controlling differentiation and genome integrity, may regulate melanoma-specific antigen expression by interfering with the endolysosomal pathway, KARS1, and antigen processing and presentation. MITF also modulates the expression of coinhibitory receptors, i.e., PD-L1 and HVEM, and the production of an inflammatory secretome, which directly affects the infiltration and/or activation of the immune cells.Furthermore, MITF is also a key determinant of melanoma cell plasticity and tumor heterogeneity, which are undoubtedly one of the major hurdles for an effective immunotherapy. Finally, we briefly discuss the role of MITF in kidney cancer, where it also plays a key role, and in immune cells, establishing MITF as a central mediator in the regulation of immune responses in melanoma and other cancers.We propose that a better understanding of MITF and immune system intersections could help in the tailoring of current ICT in melanomas and pave the way for clinical benefits and long-lasting responses.
Collapse
Affiliation(s)
- Robert Ballotti
- Université Côte d'Azur, Nice, France
- Inserm, Biology and Pathologies of melanocytes, team1, Equipe labellisée Ligue 2020 and Equipe labellisée ARC 2019, Centre Méditerranéen de Médecine Moléculaire, Nice, France
| | - Yann Cheli
- Université Côte d'Azur, Nice, France
- Inserm, Biology and Pathologies of melanocytes, team1, Equipe labellisée Ligue 2020 and Equipe labellisée ARC 2019, Centre Méditerranéen de Médecine Moléculaire, Nice, France
| | - Corine Bertolotto
- Université Côte d'Azur, Nice, France.
- Inserm, Biology and Pathologies of melanocytes, team1, Equipe labellisée Ligue 2020 and Equipe labellisée ARC 2019, Centre Méditerranéen de Médecine Moléculaire, Nice, France.
| |
Collapse
|
41
|
Annie L, Gurusubramanian G, Roy VK. Changes in the localization of ovarian visfatin protein and its possible role during estrous cycle of mice. Acta Histochem 2020; 122:151630. [PMID: 32992122 DOI: 10.1016/j.acthis.2020.151630] [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: 07/01/2020] [Revised: 09/15/2020] [Accepted: 09/15/2020] [Indexed: 12/19/2022]
Abstract
Visfatin is a crucial adipokine, which also regulates ovarian functions in many animals. Mice estrous cycle is characterized by a dynamic complex physiological process in the reproductive system. Expression of various factors changes during the estrous cycle in the ovary. To the best of our knowledge, no previous study has been conducted on the expression of visfatin in mice ovaries during the estrous cycle. Therefore, we investigated the localization and expression of visfatin protein in the ovary of mice during the estrous cycle. Western blot analysis showed the elevated expression of visfatin in proestrus and lowest in diestrus. Immunohistochemical localization of visfatin showed intense staining in the corpus luteum of proestrus and diestrus ovaries. Thecal cells, granulosa cells, and oocytes also showed the presence of visfatin. Expression of ovarian visfatin was correlated to BCL2 and active caspase3 expression and exhibited a significant positive correlation. Furthermore, in vivo inhibition of visfatin by FK866 in the proestrus ovary down-regulated active caspase3 and PCNA expression, and up-regulated the BCL2 expression. These results suggest the role of visfatin in the proliferation and apoptosis of the follicles and specific localization of visfatin in the corpus luteum also indicate its role in corpus luteum function, which may be in progesterone biosynthesis and regression of old corpus luteum. However, further study is required to support these findings. In conclusion, visfatin may also be regulating follicular growth during the estrous cycle by regulating proliferation and apoptosis.
Collapse
|
42
|
Annie L, Gurusubramanian G, Roy VK. Inhibition of visfatin/NAMPT affects ovarian proliferation, apoptosis, and steroidogenesis in pre-pubertal mice ovary. J Steroid Biochem Mol Biol 2020; 204:105763. [PMID: 32987128 DOI: 10.1016/j.jsbmb.2020.105763] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 09/18/2020] [Accepted: 09/19/2020] [Indexed: 12/15/2022]
Abstract
Pubertal ovarian function might be dependent on the factors present in the pre-pubertal stages. Visfatin regulates ovarian steroidogenesis in adult. To date, no study has investigated the role of visfatin either in pre-pubertal or pubertal mice ovary. Thus, we investigated the role of visfatin in pre-pubertal mice ovary in relation to steroidogenesis and proliferation and apoptosis in vitro by inhibiting the endogenous visfatin by a specific inhibitor, FK866. Inhibition of visfatin increased the estrogen secretion and also up-regulated the expression of CYP11A1, 17βHSD and CYP19A1 in mice ovary. Furthermore, active caspase3 was up-regulated along with the down-regulation of BAX and BCL2 in the pre-pubertal ovary after visfatin inhibition. The expression of GCNA, PCNA, and BrdU labeling was also decreased by FK866 treatment. These results suggest that visfatin inhibits steroidogenesis, increases proliferation, and suppresses apoptosis in the pre-pubertal mice ovary. So, visfatin is a new regulator of ovary function in pre-pubertal mice.
Collapse
Affiliation(s)
| | | | - Vikas Kumar Roy
- Department of Zoology, Mizoram University, Aizawl, Mizoram 796 004, India.
| |
Collapse
|
43
|
Tang Y, Durand S, Dalle S, Caramel J. EMT-Inducing Transcription Factors, Drivers of Melanoma Phenotype Switching, and Resistance to Treatment. Cancers (Basel) 2020; 12:E2154. [PMID: 32759677 PMCID: PMC7465730 DOI: 10.3390/cancers12082154] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 07/31/2020] [Accepted: 08/01/2020] [Indexed: 01/06/2023] Open
Abstract
Transcription factors, extensively described for their role in epithelial-mesenchymal transition (EMT-TFs) in epithelial cells, also display essential functions in the melanocyte lineage. Recent evidence has shown specific expression patterns and functions of these EMT-TFs in neural crest-derived melanoma compared to carcinoma. Herein, we present an update of the specific roles of EMT-TFs in melanocyte differentiation and melanoma progression. As major regulators of phenotype switching between differentiated/proliferative and neural crest stem cell-like/invasive states, these factors appear as major drivers of intra-tumor heterogeneity and resistance to treatment in melanoma, which opens new avenues in terms of therapeutic targeting.
Collapse
Affiliation(s)
- Yaqi Tang
- Cancer Cell Plasticity in Melanoma Laboratory, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, 69008 Lyon, France; (Y.T.); (S.D.); (S.D.)
| | - Simon Durand
- Cancer Cell Plasticity in Melanoma Laboratory, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, 69008 Lyon, France; (Y.T.); (S.D.); (S.D.)
| | - Stéphane Dalle
- Cancer Cell Plasticity in Melanoma Laboratory, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, 69008 Lyon, France; (Y.T.); (S.D.); (S.D.)
- Dermatology Unit, Hospices Civils de Lyon, Centre Hospitalier Lyon Sud, 69495 Pierre Bénite, France
| | - Julie Caramel
- Cancer Cell Plasticity in Melanoma Laboratory, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, 69008 Lyon, France; (Y.T.); (S.D.); (S.D.)
| |
Collapse
|
44
|
Mpilla GB, Philip PA, El-Rayes B, Azmi AS. Pancreatic neuroendocrine tumors: Therapeutic challenges and research limitations. World J Gastroenterol 2020; 26:4036-4054. [PMID: 32821069 PMCID: PMC7403797 DOI: 10.3748/wjg.v26.i28.4036] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/10/2020] [Accepted: 07/16/2020] [Indexed: 02/06/2023] Open
Abstract
Pancreatic neuroendocrine tumors (PNETs) are known to be the second most common epithelial malignancy of the pancreas. PNETs can be listed among the slowest growing as well as the fastest growing human cancers. The prevalence of PNETs is deceptively low; however, its incidence has significantly increased over the past decades. According to the American Cancer Society's estimate, about 4032 (> 7% of all pancreatic malignancies) individuals will be diagnosed with PNETs in 2020. PNETs often cause severe morbidity due to excessive secretion of hormones (such as serotonin) and/or overall tumor mass. Patients can live for many years (except for those patients with poorly differentiated G3 neuroendocrine tumors); thus, the prevalence of the tumors that is the number of patients actually dealing with the disease at any given time is fairly high because the survival is much longer than pancreatic ductal adenocarcinoma. Due to significant heterogeneity, the management of PNETs is very complex and remains an unmet clinical challenge. In terms of research studies, modest improvements have been made over the past decades in the identification of potential oncogenic drivers in order to enhance the quality of life and increase survival for this growing population of patients. Unfortunately, the majority of systematic therapies approved for the management of advanced stage PNETs lack objective response or at most result in modest benefits in survival. In this review, we aim to discuss the broad challenges associated with the management and the study of PNETs.
Collapse
Affiliation(s)
- Gabriel Benyomo Mpilla
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, United States
| | - Philip Agop Philip
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, United States
| | - Bassel El-Rayes
- Department of Hematology Oncology, Emory Winship Institute, Atlanta, GA 30322, United States
| | - Asfar Sohail Azmi
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, United States
| |
Collapse
|
45
|
Nampt-mediated spindle sizing secures a post-anaphase increase in spindle speed required for extreme asymmetry. Nat Commun 2020; 11:3393. [PMID: 32636388 PMCID: PMC7341875 DOI: 10.1038/s41467-020-17088-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 06/09/2020] [Indexed: 12/21/2022] Open
Abstract
Meiotic divisions in oocytes are extremely asymmetric and require pre- and post-anaphase-onset phases of spindle migration. The latter induces membrane protrusion that is moulded around the spindle thereby reducing cytoplasmic loss. Here, we find that depleting the NAD biosynthetic enzyme, nicotinamide phosphoribosyl-transferase (Nampt), in mouse oocytes results in markedly longer spindles and compromises asymmetry. By analysing spindle speed in live oocytes, we identify a striking and transient acceleration after anaphase-onset that is severely blunted following Nampt-depletion. Slow-moving midzones of elongated spindles induce cortical furrowing deep within the oocyte before protrusions can form, altogether resulting in larger oocyte fragments being cleaved off. Additionally, we find that Nampt-depletion lowers NAD and ATP levels and that reducing NAD using small molecule Nampt inhibitors also compromises asymmetry. These data show that rapid midzone displacement is critical for extreme asymmetry by delaying furrowing to enable protrusions to form and link metabolic status to asymmetric division. Meiotic cell division in oocytes is asymmetric and requires microtubule spindle migration after anaphase-onset. Here, the authors show that Nampt, an enzyme of the Nicotinamide adenine dinucleotide (NAD) biosynthetic pathway, contributes to post-anaphase spindle migration and oocyte division asymmetry by controlling spindle length.
Collapse
|
46
|
Gaudel C, Soysouvanh F, Leclerc J, Bille K, Husser C, Montcriol F, Bertolotto C, Ballotti R. Regulation of Melanogenesis by the Amino Acid Transporter SLC7A5. J Invest Dermatol 2020; 140:2253-2259.e4. [PMID: 32240722 DOI: 10.1016/j.jid.2020.03.941] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/26/2020] [Accepted: 03/09/2020] [Indexed: 10/24/2022]
Abstract
Integration of chromatin immunoprecipitation-sequencing and microarray data enabled us to identify previously unreported MITF-target genes, among which the amino acid transporter SLC7A5 is also included. We reported that small interfering RNA-mediated SLC7A5 knockdown decreased pigmentation in B16F10 cells but neither affected morphology nor dendricity. Treatment with the SLC7A5 inhibitors 2-amino-2-norbornanecarboxylic acid (BCH) or JPH203 also decreased melanin synthesis in B16F10 cells. Our findings indicated that BCH was as potent as reference depigmenting agent, kojic acid, but acted through a different pathway not affecting tyrosinase activity. BCH also decreased pigmentation in human MNT1 melanoma cells or normal human melanocytes. Finally, we tested BCH on a more physiological model, using reconstructed human epidermis and confirmed a strong inhibition of pigmentation, revealing the clinical potential of SLC7A5 inhibition and positioning BCH as a depigmenting agent suitable for cosmetic or dermatological intervention in hyperpigmentation diseases.
Collapse
Affiliation(s)
- Céline Gaudel
- Nunii Laboratoire, Grasse Biotech, Grasse, France; Université Nice Côte d'Azur, C3M, INSERM, U1065, Biology and pathologies of melanocytes, Nice, France
| | - Frédéric Soysouvanh
- Université Nice Côte d'Azur, C3M, INSERM, U1065, Biology and pathologies of melanocytes, Nice, France
| | - Justine Leclerc
- Université Nice Côte d'Azur, C3M, INSERM, U1065, Biology and pathologies of melanocytes, Nice, France
| | - Karine Bille
- Université Nice Côte d'Azur, C3M, INSERM, U1065, Biology and pathologies of melanocytes, Nice, France
| | - Chrystel Husser
- Université Nice Côte d'Azur, C3M, INSERM, U1065, Biology and pathologies of melanocytes, Nice, France
| | | | - Corine Bertolotto
- Université Nice Côte d'Azur, C3M, INSERM, U1065, Biology and pathologies of melanocytes, Nice, France
| | - Robert Ballotti
- Université Nice Côte d'Azur, C3M, INSERM, U1065, Biology and pathologies of melanocytes, Nice, France.
| |
Collapse
|
47
|
Silencing of NAMPT leads to up-regulation of insulin receptor substrate 1 gene expression in U87 glioma cells. Endocr Regul 2020; 54:31-42. [DOI: 10.2478/enr-2020-0005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Abstract
Objective. The aim of the present study was to investigate the effect of adipokine NAMPT (nicotinamide phosphoribosyltransferase) silencing on the expression of genes encoding IRS1 (insulin receptor substrate 1) and some other proliferation related proteins in U87 glioma cells for evaluation of the possible significance of this adipokine in intergenic interactions.
Methods. The silencing of NAMPT mRNA was introduced by NAMPT specific siRNA. The expression level of NAMPT, IGFBP3, IRS1, HK2, PER2, CLU, BNIP3, TPD52, GADD45A, and MKI67 genes was studied in U87 glioma cells by quantitative polymerase chain reaction. Anti-visfatin antibody was used for detection of NAMPT protein by Western-blot analysis.
Results. It was shown that the silencing of NAMPT mRNA led to a strong down-regulation of NAMPT protein and significant modification of the expression of IRS1, IGFBP3, CLU, HK2, BNIP3, and MKI67 genes in glioma cells and a strong up-regulation of IGFBP3 and IRS1 and down-regulation of CLU, BNIP3, HK2, and MKI67 gene expressions. At the same time, no significant changes were detected in the expression of GADD45A, PER2, and TPD52 genes in glioma cells treated by siRNA specific to NAMPT. Furthermore, the silencing of NAMPT mRNA suppressed the glioma cell proliferation.
Conclusions. Results of this investigation demonstrated that silencing of NAMPT mRNA with corresponding down-regulation of NAMPT protein and suppression of the glioma cell proliferation affected the expression of IRS1 gene as well as many other genes encoding the proliferation related proteins. It is possible that dysregulation of most of the studied genes in glioma cells after silencing of NAMPT is reflected by a complex of intergenic interactions and that NAMPT is an important factor for genome stability and regulatory mechanisms contributing to the control of glioma cell metabolism and proliferation.
Collapse
|
48
|
Ye C, Qi L, Li X, Wang J, Yu J, Zhou B, Guo C, Chen J, Zheng S. Targeting the NAD + salvage pathway suppresses APC mutation-driven colorectal cancer growth and Wnt/β-catenin signaling via increasing Axin level. Cell Commun Signal 2020; 18:16. [PMID: 32005247 PMCID: PMC6995173 DOI: 10.1186/s12964-020-0513-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 01/17/2020] [Indexed: 02/05/2023] Open
Abstract
Background The role and mechanism of the nicotinamide adenine dinucleotide (NAD+) salvage pathway in cancer cell proliferation is poorly understood. Nicotinamide phosphoribosyltransferase (NAMPT), which converts nicotinamide into NAD+, is the rate-limiting enzyme in the NAD+ salvage pathway. Here, we assessed the role of NAMPT in the proliferation of colorectal cancer. Methods Real-time PCR, immunohistochemistry, western blotting, and analyses of datasets from Oncomine and Gene Expression Omnibus were conducted to assess the expression of NAMPT at the mRNA and protein levels in colorectal cancer. The Kaplan Meier plotter online tool was used to evaluate the prognostic role of NAMPT. Knockdown of NAMPT was performed to assess the role of NAMPT in colorectal cancer cell proliferation and tumorigenesis both in vitro and in vivo. Overexpression of NAMPT was used to evaluate impact of NAMPT on colorectal cancer cell proliferation in vitro. NAD+ quantitation, immunofluorescence, dual luciferase assay and western blot were used to explore the mechanism of colorectal cancer proliferation. Transwell migration and invasion assays were conducted to assess the role of NAMPT in cell migration and invasion abilities of colorectal cancer cells. Results Our study indicated that the inhibition of NAMPT decreased proliferation capacity of colorectal cancer cells both in vitro and in vivo. Conversely, overexpression of NAMPT could promote cell proliferation in vitro. NAMPT inhibition induced β-catenin degradation by increasing Axin expression levels; this resulted in the inhibition of Wnt/β-catenin signaling and cell proliferation in colorectal cancer. The addition of nicotinamide mononucleotide, the enzymatic product of NAMPT, effectively reversed β-catenin protein degradation and inhibited growth. Similarly, the knockdown of Axin also decreased the cell death induced by the inhibition of NAMPT. In addition, we showed that colorectal cancer tissues harbored significantly higher levels of NAMPT than the levels harbored by paired normal tissues, especially in colorectal cancer stages I and II. And the overexpression of NAMPT was associated with unfavorable survival results. Conclusions Our findings reveal that NAMPT plays an important role in colorectal cancer proliferation via Wnt/β-catenin pathway, which could have vital implications for the diagnosis, prognosis and treatment of colorectal cancer.
Collapse
Affiliation(s)
- Chenyang Ye
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), the Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, 310009, Hangzhou, China
| | - Lina Qi
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), the Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, 310009, Hangzhou, China
| | - Xiaofen Li
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), the Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, 310009, Hangzhou, China.,Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ji Wang
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Zhejiang, 310016, Hangzhou, China.,Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Zhejiang, 310016, Hangzhou, China
| | - Jiekai Yu
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), the Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, 310009, Hangzhou, China
| | - Biting Zhou
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), the Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, 310009, Hangzhou, China
| | - Cheng Guo
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), the Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, 310009, Hangzhou, China
| | - Jiani Chen
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), the Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, 310009, Hangzhou, China
| | - Shu Zheng
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), the Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, 310009, Hangzhou, China. .,Reseach Center for Air Pollution and Health, School of Medicine, Zhejiang University, Zhejiang, 310009, Hangzhou, China.
| |
Collapse
|
49
|
Heske CM. Beyond Energy Metabolism: Exploiting the Additional Roles of NAMPT for Cancer Therapy. Front Oncol 2020; 9:1514. [PMID: 32010616 PMCID: PMC6978772 DOI: 10.3389/fonc.2019.01514] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 12/16/2019] [Indexed: 12/13/2022] Open
Abstract
Tumor cells have increased requirements for NAD+. Thus, many cancers exhibit an increased reliance on NAD+ production pathways. This dependence may be exploited therapeutically through pharmacological targeting of NAMPT, the rate-limiting enzyme in the NAD+ salvage pathway. Despite promising preclinical data using NAMPT inhibitors in cancer models, early NAMPT inhibitors showed limited efficacy in several early phase clinical trials, necessitating the identification of strategies, such as drug combinations, to enhance their efficacy. While the effect of NAMPT inhibitors on impairment of energy metabolism in cancer cells has been well-described, more recent insights have uncovered a number of additional targetable cellular processes that are impacted by inhibition of NAMPT. These include sirtuin function, DNA repair machinery, redox homeostasis, molecular signaling, cellular stemness, and immune processes. This review highlights the recent findings describing the effects of NAMPT inhibitors on the non-metabolic functions of malignant cells, with a focus on how this information can be leveraged clinically. Combining NAMPT inhibitors with other therapies that target NAD+-dependent processes or selecting tumors with specific vulnerabilities that can be co-targeted with NAMPT inhibitors may represent opportunities to exploit the multiple functions of this enzyme for greater therapeutic benefit.
Collapse
Affiliation(s)
- Christine M Heske
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
50
|
Strub T, Ballotti R, Bertolotto C. The "ART" of Epigenetics in Melanoma: From histone "Alterations, to Resistance and Therapies". Theranostics 2020; 10:1777-1797. [PMID: 32042336 PMCID: PMC6993228 DOI: 10.7150/thno.36218] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 11/14/2019] [Indexed: 02/07/2023] Open
Abstract
Malignant melanoma is the most deadly form of skin cancer. It originates from melanocytic cells and can also arise at other body sites. Early diagnosis and appropriate medical care offer excellent prognosis with up to 5-year survival rate in more than 95% of all patients. However, long-term survival rate for metastatic melanoma patients remains at only 5%. Indeed, malignant melanoma is known for its notorious resistance to most current therapies and is characterized by both genetic and epigenetic alterations. In cutaneous melanoma (CM), genetic alterations have been implicated in drug resistance, yet the main cause of this resistance seems to be non-genetic in nature with a change in transcription programs within cell subpopulations. This change can adapt and escape targeted therapy and immunotherapy cytotoxic effects favoring relapse. Because they are reversible in nature, epigenetic changes are a growing focus in cancer research aiming to prevent or revert the drug resistance with current therapies. As such, the field of epigenetic therapeutics is among the most active area of preclinical and clinical research with effects of many classes of epigenetic drugs being investigated. Here, we review the multiplicity of epigenetic alterations, mainly histone alterations and chromatin remodeling in both cutaneous and uveal melanomas, opening opportunities for further research in the field and providing clues to specifically control these modifications. We also discuss how epigenetic dysregulations may be exploited to achieve clinical benefits for the patients, the limitations of these therapies, and recent data exploring this potential through combinatorial epigenetic and traditional therapeutic approaches.
Collapse
Affiliation(s)
- Thomas Strub
- Université Nice Côte d'Azur, Inserm, C3M, France
- Biology and pathologies of melanocytes, Equipe labellisée ARC 2019, C3M, team 1, France
| | - Robert Ballotti
- Université Nice Côte d'Azur, Inserm, C3M, France
- Biology and pathologies of melanocytes, Equipe labellisée ARC 2019, C3M, team 1, France
| | - Corine Bertolotto
- Université Nice Côte d'Azur, Inserm, C3M, France
- Biology and pathologies of melanocytes, Equipe labellisée ARC 2019, C3M, team 1, France
| |
Collapse
|