1
|
John J, Das S, Kunnath A, Mudgal J, Nandakumar K. Effects of quercetin and derivatives on NAMPT/Sirtuin-1 metabolic pathway in neuronal cells: an approach to mitigate chemotherapy-induced cognitive impairment. Metab Brain Dis 2025; 40:151. [PMID: 40085284 PMCID: PMC11909064 DOI: 10.1007/s11011-025-01567-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 03/02/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND The cognitive alterations observed in individuals undergoing cancer treatments have garnered more attention recently. Chemotherapy can reduce nicotinamide adenine dinucleotide (NAD+) levels by inhibiting nicotinamide phosphoribosyl transferase (NAMPT). This reduction can make cancer cells more susceptible to oxidative damage and death and may also affect non-cancerous cells, particularly the brain cells. During chemotherapy-induced suppression, the downregulation of the NAMPT-mediated NAD+/Sirtuin 1 (SIRT1) pathway may cause dyscognition. Objective: This study aimed to assess the role of quercetin and analogues in chemobrain and the associated mechanisms. Methods: The potential of quercetin and its derivatives interaction with NAMPT and SIRT1 proteins was performed using computational studies followed by their in vitro evaluation in SH-SY5Y cells. Molecular docking and simulation studies of human SIRT1 and NAMPT proteins with quercetin and its derivatives were performed. Differentiated SH-SY5Y cell lines were treated with quercetin and selected derivatives against Methotrexate and 5-Fluorouracil (MF) toxicity, by subjecting to cytotoxicity assay, flow cytometry, and RT-PCR analysis. Results: Quercetin, Rutin, and Isoquercetin showed interactions necessary in the activation process of both proteins. Cytotoxicity and flow cytometric studies demonstrated that the phytochemicals shield the differentiated SH-SY5Y cells from MF toxicity. As determined by RT-PCR investigations, NAMPT and SIRT1 gene mRNA expression was higher in test drug-treated cells at quercetin (0.12, 0.6 µM), rutin, and isoquercetin (16, 80 µM) and lower in MF-treated cells. Conclusion: The treatment of phytochemicals alleviated CICI by targeting NAMPT and SIRT1 proteins, which could lead to the identification of effective treatment strategies for the chemobrain.
Collapse
Affiliation(s)
- Jeena John
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Subham Das
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Anu Kunnath
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Jayesh Mudgal
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
- School of Pharmaceutical Sciences, Manipal University Jaipur, Jaipur, 303007, Rajasthan, India
| | - Krishnadas Nandakumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
- Centre for Animal Research, Ethics and Training, Manipal Academy of Higher Education, Manipal, India.
| |
Collapse
|
2
|
Ozgencil F, Gunindi HB, Eren G. Dual-targeted NAMPT inhibitors as a progressive strategy for cancer therapy. Bioorg Chem 2024; 149:107509. [PMID: 38824699 DOI: 10.1016/j.bioorg.2024.107509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 04/29/2024] [Accepted: 05/28/2024] [Indexed: 06/04/2024]
Abstract
In mammals, nicotinamide phosphoribosyltransferase (NAMPT) is a crucial enzyme in the nicotinamide adenine dinucleotide (NAD+) synthesis pathway catalyzing the condensation of nicotinamide (NAM) with 5-phosphoribosyl-1-pyrophosphate (PRPP) to produce nicotinamide mononucleotide (NMN). Given the pivotal role of NAD+ in a range of cellular functions, including DNA synthesis, redox reactions, cytokine generation, metabolism, and aging, NAMPT has become a promising target for many diseases, notably cancer. Therefore, various NAMPT inhibitors have been reported and classified as first and second-generation based on their chemical structures and design strategies, dual-targeted being one. However, most NAMPT inhibitors suffer from several limitations, such as dose-dependent toxicity and poor pharmacokinetic properties. Consequently, there is no clinically approved NAMPT inhibitor. Hence, research on discovering more effective and less toxic dual-targeted NAMPT inhibitors with desirable pharmacokinetic properties has drawn attention recently. This review summarizes the previously reported dual-targeted NAMPT inhibitors, focusing on their design strategies and advantages over the single-targeted therapies.
Collapse
Affiliation(s)
- Fikriye Ozgencil
- SIRTeam Group, Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, 06330 Ankara, Türkiye
| | - Habibe Beyza Gunindi
- SIRTeam Group, Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, 06330 Ankara, Türkiye
| | - Gokcen Eren
- SIRTeam Group, Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, 06330 Ankara, Türkiye.
| |
Collapse
|
3
|
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
|
4
|
Zhu X, Liu H, Chen L, Wu C, Liu X, Cang Y, Jiang B, Yang X, Fan G. Addressing the Enzyme-independent tumor-promoting function of NAMPT via PROTAC-mediated degradation. Cell Chem Biol 2022; 29:1616-1629.e12. [PMID: 36323324 DOI: 10.1016/j.chembiol.2022.10.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 08/28/2022] [Accepted: 10/10/2022] [Indexed: 11/07/2022]
Abstract
Aberrant overexpression of nicotinamide phosphoribosyltransferase (NAMPT) has been reported in a variety of tumor cells and is a poor prognosis factor for patient survival. It plays an important role in tumor cell proliferation, acting concurrently as an nicotinamide adenine dinucleotide (NAD+) synthase and, unexpectedly, as an extracellular signaling molecule for several tumor-promoting pathways. Although previous efforts to modulate NAMPT activity were limited to enzymatic inhibitors with low success in clinical studies, protein degradation offers the possibility to simultaneously disrupt NAMPT's enzyme activity and ligand capabilities. Here we report the development of two highly selective proteolysis-targeting chimeras (PROTACs) that promote NAMPT degradation in a cereblon-dependent manner. Both PROTAC degraders outperform a clinical candidate, FK866, in killing effect on hematological tumor cells. These results emphasize the importance and feasibility of applying PROTACs as a superior strategy for targeting proteins with multiple tumor-promoting functions like NAMPT, which is not easily achieved by conventional enzymatic inhibitors.
Collapse
Affiliation(s)
- Xiaotong Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Haixia Liu
- School of Physical Science and Technology, ShanghaiTech University, Shanghai 201210, China; Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Li Chen
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Chenxu Wu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xuesong Liu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yong Cang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Biao Jiang
- CAS Key Laboratory of Synthetic Chemistry of Natural Substances, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China.
| | - Xiaobao Yang
- Gluetacs Therapeutics (Shanghai) Co., Ltd., Zhangjiang Hi-Tech Park, Shanghai 201210, China.
| | - Gaofeng Fan
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
5
|
Sun D, Zhang J, Dong G, He S, Sheng C. Blocking Non-enzymatic Functions by PROTAC-Mediated Targeted Protein Degradation. J Med Chem 2022; 65:14276-14288. [DOI: 10.1021/acs.jmedchem.2c01159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Donghuan Sun
- School of Pharmacy, Second Military Medical University (Naval Medical University), Shanghai 200433, China
| | - Jing Zhang
- Department of Pathology, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai 200003, China
| | - Guoqiang Dong
- School of Pharmacy, Second Military Medical University (Naval Medical University), Shanghai 200433, China
| | - Shipeng He
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
| | - Chunquan Sheng
- School of Pharmacy, Second Military Medical University (Naval Medical University), Shanghai 200433, China
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Xu Q, Liu X, Mohseni G, Hao X, Ren Y, Xu Y, Gao H, Wang Q, Wang Y. Mechanism research and treatment progress of NAD pathway related molecules in tumor immune microenvironment. Cancer Cell Int 2022; 22:242. [PMID: 35906622 PMCID: PMC9338646 DOI: 10.1186/s12935-022-02664-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/19/2022] [Indexed: 11/21/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD) is the core of cellular energy metabolism. NAMPT, Sirtuins, PARP, CD38, and other molecules in this classic metabolic pathway affect many key cellular functions and are closely related to the occurrence and development of many diseases. In recent years, several studies have found that these molecules can regulate cell energy metabolism, promote the release of related cytokines, induce the expression of neoantigens, change the tumor immune microenvironment (TIME), and then play an anticancer role. Drugs targeting these molecules are under development or approved for clinical use. Although there are some side effects and drug resistance, the discovery of novel drugs, the development of combination therapies, and the application of new technologies provide solutions to these challenges and improve efficacy. This review presents the mechanisms of action of NAD pathway-related molecules in tumor immunity, advances in drug research, combination therapies, and some new technology-related therapies.
Collapse
Affiliation(s)
- QinChen Xu
- Department of Clinical Laboratory, The Second Hospital of Shandong University, 247 Beiyuan Street, 250033, Jinan, Shandong, China
| | - Xiaoyan Liu
- Department of Clinical Laboratory, The Second Hospital of Shandong University, 247 Beiyuan Street, 250033, Jinan, Shandong, China
| | - Ghazal Mohseni
- Department of Clinical Laboratory, The Second Hospital of Shandong University, 247 Beiyuan Street, 250033, Jinan, Shandong, China
| | - Xiaodong Hao
- Department of Clinical Laboratory, The Second Hospital of Shandong University, 247 Beiyuan Street, 250033, Jinan, Shandong, China
| | - Yidan Ren
- Department of Clinical Laboratory, The Second Hospital of Shandong University, 247 Beiyuan Street, 250033, Jinan, Shandong, China
| | - Yiwei Xu
- Marine College, Shandong University, 264209, Weihai, China
| | - Huiru Gao
- Department of Clinical Laboratory, The Second Hospital of Shandong University, 247 Beiyuan Street, 250033, Jinan, Shandong, China
| | - Qin Wang
- Department of Anesthesiology, Cheeloo College of Medicine, Qilu Hospital, Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong, China.
| | - Yunshan Wang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, 247 Beiyuan Street, 250033, Jinan, Shandong, China.
| |
Collapse
|
9
|
Curry A, White D, Cen Y. Small Molecule Regulators Targeting NAD + Biosynthetic Enzymes. Curr Med Chem 2022; 29:1718-1738. [PMID: 34060996 PMCID: PMC8630097 DOI: 10.2174/0929867328666210531144629] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 04/02/2021] [Accepted: 04/07/2021] [Indexed: 01/03/2023]
Abstract
Nicotinamide adenine dinucleotide (NAD+) is a key player in many metabolic pathways as an activated carrier of electrons. In addition to being the cofactor for redox reactions, NAD+ also serves as the substrate for various enzymatic transformations such as adenylation and ADP-ribosylation. Maintaining cellular NAD+ homeostasis has been suggested as an effective anti-aging strategy. Given the importance of NAD+ in regulating a broad spectrum of cellular events, small molecules targeting NAD+ metabolism have been pursued as therapeutic interventions for the treatment of mitochondrial disorders and agerelated diseases. In this article, small molecule regulators of NAD+ biosynthetic enzymes will be reviewed. The focus will be given to the discovery and development of these molecules, the mechanism of action as well as their therapeutic potentials.
Collapse
Affiliation(s)
- Alyson Curry
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23219, USA
| | - Dawanna White
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23219, USA
| | - Yana Cen
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23219, USA;,Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA 23219, USA,Address correspondence to this author at the Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23219, USA; Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA 23219, USA; Tel: 804-828-7405;
| |
Collapse
|
10
|
Roussin M, Salcedo SP. NAD+-targeting by bacteria: an emerging weapon in pathogenesis. FEMS Microbiol Rev 2021; 45:6315328. [PMID: 34223888 DOI: 10.1093/femsre/fuab037] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 07/01/2021] [Indexed: 11/14/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) is a major cofactor in redox reactions in all lifeforms. A stable level of NAD+ is vital to ensure cellular homeostasis. Some pathogens can modulate NAD+ metabolism to their advantage and even utilize or cleave NAD+ from the host using specialized effectors known as ADP-ribosyltransferase toxins and NADases, leading to energy store depletion, immune evasion, or even cell death. This review explores recent advances in the field of bacterial NAD+-targeting toxins, highlighting the relevance of NAD+ modulation as an emerging pathogenesis strategy. In addition, we discuss the role of specific NAD+-targeting toxins in niche colonization and bacterial lifestyle as components of Toxin/Antitoxin systems and key players in inter-bacterial competition. Understanding the mechanisms of toxicity, regulation, and secretion of these toxins will provide interesting leads in the search for new antimicrobial treatments in the fight against infectious diseases.
Collapse
Affiliation(s)
- Morgane Roussin
- Laboratory of Molecular Microbiology and Structural Biochemistry, Centre National de la Recherche Scientifique UMR5086, Université de Lyon, Lyon, France
| | - Suzana P Salcedo
- Laboratory of Molecular Microbiology and Structural Biochemistry, Centre National de la Recherche Scientifique UMR5086, Université de Lyon, Lyon, France
| |
Collapse
|
11
|
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
|
12
|
Ghanem MS, Monacelli F, Nencioni A. Advances in NAD-Lowering Agents for Cancer Treatment. Nutrients 2021; 13:1665. [PMID: 34068917 PMCID: PMC8156468 DOI: 10.3390/nu13051665] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/04/2021] [Accepted: 05/08/2021] [Indexed: 12/13/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD) is an essential redox cofactor, but it also acts as a substrate for NAD-consuming enzymes, regulating cellular events such as DNA repair and gene expression. Since such processes are fundamental to support cancer cell survival and proliferation, sustained NAD production is a hallmark of many types of neoplasms. Depleting intratumor NAD levels, mainly through interference with the NAD-biosynthetic machinery, has emerged as a promising anti-cancer strategy. NAD can be generated from tryptophan or nicotinic acid. In addition, the "salvage pathway" of NAD production, which uses nicotinamide, a byproduct of NAD degradation, as a substrate, is also widely active in mammalian cells and appears to be highly exploited by a subset of human cancers. In fact, research has mainly focused on inhibiting the key enzyme of the latter NAD production route, nicotinamide phosphoribosyltransferase (NAMPT), leading to the identification of numerous inhibitors, including FK866 and CHS-828. Unfortunately, the clinical activity of these agents proved limited, suggesting that the approaches for targeting NAD production in tumors need to be refined. In this contribution, we highlight the recent advancements in this field, including an overview of the NAD-lowering compounds that have been reported so far and the related in vitro and in vivo studies. We also describe the key NAD-producing pathways and their regulation in cancer cells. Finally, we summarize the approaches that have been explored to optimize the therapeutic response to NAMPT inhibitors in cancer.
Collapse
Affiliation(s)
- Moustafa S. Ghanem
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, Viale Benedetto XV 6, 16132 Genoa, Italy; (M.S.G.); (F.M.)
| | - Fiammetta Monacelli
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, Viale Benedetto XV 6, 16132 Genoa, Italy; (M.S.G.); (F.M.)
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Alessio Nencioni
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, Viale Benedetto XV 6, 16132 Genoa, Italy; (M.S.G.); (F.M.)
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132 Genova, Italy
| |
Collapse
|
13
|
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
|
14
|
Selective targeting of NAMPT by KPT-9274 in acute myeloid leukemia. Blood Adv 2020; 3:242-255. [PMID: 30692102 DOI: 10.1182/bloodadvances.2018024182] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Accepted: 12/06/2018] [Indexed: 12/30/2022] Open
Abstract
Treatment options for acute myeloid leukemia (AML) remain extremely limited and associated with significant toxicity. Nicotinamide phosphoribosyltransferase (NAMPT) is involved in the generation of NAD+ and a potential therapeutic target in AML. We evaluated the effect of KPT-9274, a p21-activated kinase 4/NAMPT inhibitor that possesses a unique NAMPT-binding profile based on in silico modeling compared with earlier compounds pursued against this target. KPT-9274 elicited loss of mitochondrial respiration and glycolysis and induced apoptosis in AML subtypes independent of mutations and genomic abnormalities. These actions occurred mainly through the depletion of NAD+, whereas genetic knockdown of p21-activated kinase 4 did not induce cytotoxicity in AML cell lines or influence the cytotoxic effect of KPT-9274. KPT-9274 exposure reduced colony formation, increased blast differentiation, and diminished the frequency of leukemia-initiating cells from primary AML samples; KPT-9274 was minimally cytotoxic toward normal hematopoietic or immune cells. In addition, KPT-9274 improved overall survival in vivo in 2 different mouse models of AML and reduced tumor development in a patient-derived xenograft model of AML. Overall, KPT-9274 exhibited broad preclinical activity across a variety of AML subtypes and warrants further investigation as a potential therapeutic agent for AML.
Collapse
|
15
|
Lv R, Yu J, Sun Q. Anti-angiogenic role of microRNA-23b in melanoma by disturbing NF-κB signaling pathway via targeted inhibition of NAMPT. Future Oncol 2020; 16:541-458. [PMID: 32107941 DOI: 10.2217/fon-2019-0699] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Aim: Melanoma is the major cause of death in patients inflicting skin cancer. We identify miR-23b plays an anti-angiogenic role in melanoma. Materials & methods: We collected tumor tissues from melanoma patients. Experiments in vivo and in vitro were designed to evaluate the role of miR-23b in melanoma. Results & conclusion: miR-23b was found to be downregulated in melanoma tissues, and associated with poor patient survival. Elevating miR-23b inhibited cell viability and colony formation, reduced pro-angiogenetic ability, and accelerated apoptosis in SK-MEL-28 cells. miR-23b targeted NAMPT. Disturbing NF-κB signaling pathway with ammonium pyrrolidinedithiocarbamate (an inhibitor of NF-kB signaling pathway) impeded acquired pro-angiogenetic ability of nicotinamide phosphoribosyl transferase-overexpressed SK-MEL-28 cells. MiR-23b is a prognostic factor in melanoma. This study provides an enhanced understanding of microRNA-based targets for melanoma treatment.
Collapse
Affiliation(s)
- Renrong Lv
- Department of Burn & Plastic Surgery, Provincial Hospital Affiliated to Shandong University, Ji'nan 250021, Shandong Province, PR China
| | - Jing Yu
- Department of Burn & Plastic Surgery, Zhangqiu People's Hospital, Ji'nan 250200, Shandong Province, PR China
| | - Qian Sun
- Department of Obstetrics, Ji'nan Maternity & Child Health Care Hospital, Ji'nan 250001, Shandong Province, PR China
| |
Collapse
|
16
|
Torretta S, Colombo G, Travelli C, Boumya S, Lim D, Genazzani AA, Grolla AA. The Cytokine Nicotinamide Phosphoribosyltransferase (eNAMPT; PBEF; Visfatin) Acts as a Natural Antagonist of C-C Chemokine Receptor Type 5 (CCR5). Cells 2020; 9:cells9020496. [PMID: 32098202 PMCID: PMC7072806 DOI: 10.3390/cells9020496] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 02/15/2020] [Accepted: 02/21/2020] [Indexed: 12/23/2022] Open
Abstract
(1) Background: Extracellular nicotinamide phosphoribosyltrasferase (eNAMPT) is released by various cell types with pro-tumoral and pro-inflammatory properties. In cancer, eNAMPT regulates tumor growth through the activation of intracellular pathways, suggesting that it acts through a putative receptor, although its nature is still elusive. It has been shown, using surface plasma resonance, that eNAMPT binds to the C-C chemokine receptor type 5 (CCR5), although the physiological meaning of this finding is unknown. The aim of the present work was to characterize the pharmacodynamics of eNAMPT on CCR5. (2) Methods: HeLa CCR5-overexpressing stable cell line and B16 melanoma cells were used. We focused on some phenotypic effects of CCR5 activation, such as calcium release and migration, to evaluate eNAMPT actions on this receptor. (3) Results: eNAMPT did not induce ERK activation or cytosolic Ca2+-rises alone. Furthermore, eNAMPT prevents CCR5 internalization mediated by Rantes. eNAMPT pretreatment inhibits CCR5-mediated PKC activation and Rantes-dependent calcium signaling. The effect of eNAMPT on CCR5 was specific, as the responses to ATP and carbachol were unaffected. This was strengthened by the observation that eNAMPT inhibited Rantes-induced Ca2+-rises and Rantes-induced migration in a melanoma cell line. (4) Conclusions: Our work shows that eNAMPT binds to CCR5 and acts as a natural antagonist of this receptor.
Collapse
Affiliation(s)
- Simone Torretta
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, 28100 Novara, Italy; (S.T.); (G.C.); (S.B.); (D.L.); (A.A.G.)
| | - Giorgia Colombo
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, 28100 Novara, Italy; (S.T.); (G.C.); (S.B.); (D.L.); (A.A.G.)
| | - Cristina Travelli
- Department of Pharmaceutical Sciences, Università di Pavia, 27100 Pavia, Italy;
| | - Sara Boumya
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, 28100 Novara, Italy; (S.T.); (G.C.); (S.B.); (D.L.); (A.A.G.)
| | - Dmitry Lim
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, 28100 Novara, Italy; (S.T.); (G.C.); (S.B.); (D.L.); (A.A.G.)
| | - Armando A. Genazzani
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, 28100 Novara, Italy; (S.T.); (G.C.); (S.B.); (D.L.); (A.A.G.)
| | - Ambra A. Grolla
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, 28100 Novara, Italy; (S.T.); (G.C.); (S.B.); (D.L.); (A.A.G.)
- Correspondence: ; Tel.: +39-0321-375822; Fax: +39-0321-375821
| |
Collapse
|
17
|
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
|
18
|
Grolla AA, Miggiano R, Di Marino D, Bianchi M, Gori A, Orsomando G, Gaudino F, Galli U, Del Grosso E, Mazzola F, Angeletti C, Guarneri M, Torretta S, Calabrò M, Boumya S, Fan X, Colombo G, Travelli C, Rocchio F, Aronica E, Wohlschlegel JA, Deaglio S, Rizzi M, Genazzani AA, Garavaglia S. A nicotinamide phosphoribosyltransferase-GAPDH interaction sustains the stress-induced NMN/NAD + salvage pathway in the nucleus. J Biol Chem 2020; 295:3635-3651. [PMID: 31988240 DOI: 10.1074/jbc.ra119.010571] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 01/16/2020] [Indexed: 12/26/2022] Open
Abstract
All cells require sustained intracellular energy flux, which is driven by redox chemistry at the subcellular level. NAD+, its phosphorylated variant NAD(P)+, and its reduced forms NAD(P)/NAD(P)H are all redox cofactors with key roles in energy metabolism and are substrates for several NAD-consuming enzymes (e.g. poly(ADP-ribose) polymerases, sirtuins, and others). The nicotinamide salvage pathway, constituted by nicotinamide mononucleotide adenylyltransferase (NMNAT) and nicotinamide phosphoribosyltransferase (NAMPT), mainly replenishes NAD+ in eukaryotes. However, unlike NMNAT1, NAMPT is not known to be a nuclear protein, prompting the question of how the nuclear NAD+ pool is maintained and how it is replenished upon NAD+ consumption. In the present work, using human and murine cells; immunoprecipitation, pulldown, and surface plasmon resonance assays; and immunofluorescence, small-angle X-ray scattering, and MS-based analyses, we report that GAPDH and NAMPT form a stable complex that is essential for nuclear translocation of NAMPT. This translocation furnishes NMN to replenish NAD+ to compensate for the activation of NAD-consuming enzymes by stressful stimuli induced by exposure to H2O2 or S-nitrosoglutathione and DNA damage inducers. These results indicate that by forming a complex with GAPDH, NAMPT can translocate to the nucleus and thereby sustain the stress-induced NMN/NAD+ salvage pathway.
Collapse
Affiliation(s)
- Ambra A Grolla
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Largo Donegani 2, 28100 Novara, Italy
| | - Riccardo Miggiano
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Largo Donegani 2, 28100 Novara, Italy
| | - Daniele Di Marino
- Department of Life and Environmental Sciences, New York-Marche Structural Biology Center (NY-MaSBiC), Polytechnic University of Marche, Via Brecce Bianche, 60131 Ancona, Italy
| | - Michele Bianchi
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Largo Donegani 2, 28100 Novara, Italy
| | - Alessandro Gori
- Istituto di Chimica del Riconoscimento Molecolare (ICRM-CNR), via Mario Bianco 9, 20131 Milano, Italy
| | - Giuseppe Orsomando
- Department of Clinical Sciences (DISCO), Section of Biochemistry, Polytechnic University of Marche, Via Ranieri 67, 60128 Ancona, Italy
| | - Federica Gaudino
- Department of Medical Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy
| | - Ubaldina Galli
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Largo Donegani 2, 28100 Novara, Italy
| | - Erika Del Grosso
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Largo Donegani 2, 28100 Novara, Italy
| | - Francesca Mazzola
- Department of Clinical Sciences (DISCO), Section of Biochemistry, Polytechnic University of Marche, Via Ranieri 67, 60128 Ancona, Italy
| | - Carlo Angeletti
- Department of Clinical Sciences (DISCO), Section of Biochemistry, Polytechnic University of Marche, Via Ranieri 67, 60128 Ancona, Italy
| | - Martina Guarneri
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Largo Donegani 2, 28100 Novara, Italy
| | - Simone Torretta
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Largo Donegani 2, 28100 Novara, Italy
| | - Marta Calabrò
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Largo Donegani 2, 28100 Novara, Italy
| | - Sara Boumya
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Largo Donegani 2, 28100 Novara, Italy
| | - Xiaorui Fan
- Department of Biological Chemistry, UCLA, Los Angeles, California 90095
| | - Giorgia Colombo
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Largo Donegani 2, 28100 Novara, Italy
| | - Cristina Travelli
- Department of Pharmaceutical Sciences, University of Pavia, Via Taramelli 12/14, 27100 Pavia, Italy
| | - Francesca Rocchio
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Largo Donegani 2, 28100 Novara, Italy
| | - Eleonora Aronica
- Amsterdam UMC, University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, 1001 NK Amsterdam, The Netherlands
| | | | - Silvia Deaglio
- Department of Medical Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy; Italian Institute for Genomic Medicine, Via Nizza 52, 10126 Turin, Italy
| | - Menico Rizzi
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Largo Donegani 2, 28100 Novara, Italy
| | - Armando A Genazzani
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Largo Donegani 2, 28100 Novara, Italy.
| | - Silvia Garavaglia
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Largo Donegani 2, 28100 Novara, Italy.
| |
Collapse
|
19
|
Wilk A, Hayat F, Cunningham R, Li J, Garavaglia S, Zamani L, Ferraris DM, Sykora P, Andrews J, Clark J, Davis A, Chaloin L, Rizzi M, Migaud M, Sobol RW. Extracellular NAD + enhances PARP-dependent DNA repair capacity independently of CD73 activity. Sci Rep 2020; 10:651. [PMID: 31959836 PMCID: PMC6971268 DOI: 10.1038/s41598-020-57506-9] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 12/29/2019] [Indexed: 02/06/2023] Open
Abstract
Changes in nicotinamide adenine dinucleotide (NAD+) levels that compromise mitochondrial function trigger release of DNA damaging reactive oxygen species. NAD+ levels also affect DNA repair capacity as NAD+ is a substrate for PARP-enzymes (mono/poly-ADP-ribosylation) and sirtuins (deacetylation). The ecto-5′-nucleotidase CD73, an ectoenzyme highly expressed in cancer, is suggested to regulate intracellular NAD+ levels by processing NAD+ and its bio-precursor, nicotinamide mononucleotide (NMN), from tumor microenvironments, thereby enhancing tumor DNA repair capacity and chemotherapy resistance. We therefore investigated whether expression of CD73 impacts intracellular NAD+ content and NAD+-dependent DNA repair capacity. Reduced intracellular NAD+ levels suppressed recruitment of the DNA repair protein XRCC1 to sites of genomic DNA damage and impacted the amount of accumulated DNA damage. Further, decreased NAD+ reduced the capacity to repair DNA damage induced by DNA alkylating agents. Overall, reversal of these outcomes through NAD+ or NMN supplementation was independent of CD73. In opposition to its proposed role in extracellular NAD+ bioprocessing, we found that recombinant human CD73 only poorly processes NMN but not NAD+. A positive correlation between CD73 expression and intracellular NAD+ content could not be made as CD73 knockout human cells were efficient in generating intracellular NAD+ when supplemented with NAD+ or NMN.
Collapse
Affiliation(s)
- Anna Wilk
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA.,Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL, 36604, USA
| | - Faisal Hayat
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA.,Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL, 36604, USA
| | - Richard Cunningham
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA.,Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL, 36604, USA
| | - Jianfeng Li
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA.,Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL, 36604, USA
| | - Silvia Garavaglia
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Largo Donegani 2, 28100, Novara, Italy
| | - Leila Zamani
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA
| | - Davide M Ferraris
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Largo Donegani 2, 28100, Novara, Italy
| | - Peter Sykora
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA.,Amelia Technologies, 14676 Rothgeb Drive, Rockville, MD, 20850, USA
| | - Joel Andrews
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA
| | - Jennifer Clark
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA.,Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL, 36604, USA
| | - Amanda Davis
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA
| | - Laurent Chaloin
- Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, CNRS, 34293, Montpellier, France
| | - Menico Rizzi
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Largo Donegani 2, 28100, Novara, Italy
| | - Marie Migaud
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA.,Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL, 36604, USA
| | - Robert W Sobol
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA. .,Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL, 36604, USA.
| |
Collapse
|
20
|
Gaudino F, Manfredonia I, Managò A, Audrito V, Raffaelli N, Vaisitti T, Deaglio S. Subcellular Characterization of Nicotinamide Adenine Dinucleotide Biosynthesis in Metastatic Melanoma by Using Organelle-Specific Biosensors. Antioxid Redox Signal 2019; 31:1150-1165. [PMID: 31456414 DOI: 10.1089/ars.2019.7799] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Aim: Nicotinamide adenine dinucleotide (NAD+) plays central roles in a wide array of normal and pathological conditions. Inhibition of NAD+ biosynthesis can be exploited therapeutically in cancer, including melanoma. To obtain quantitation of NAD+ levels in live cells and to address the issue of the compartmentalization of NAD+ biosynthesis, we exploited a recently described genetically encoded NAD+ biosensor (LigA-circularly permutated Venus), which was targeted to the cytosol, mitochondria, and nuclei of BRAF-V600E A375 melanoma cells, a model of metastatic melanoma (MM). Results: FK866, a specific inhibitor of nicotinamide phosphoribosyltransferase (NAMPT), the main NAD+-producing enzyme in MM cells, was used to monitor NAD+ depletion kinetics at the subcellular level in biosensor-transduced A375 cells. In addition, we treated FK866-blocked A375 cells with NAD+ precursors, including nicotinamide, nicotinic acid, nicotinamide riboside, and quinolinic acid, highlighting an organelle-specific capacity of each substrate to rescue from NAMPT block. Expression of NAD+ biosynthetic enzymes was then biochemically studied in isolated organelles, revealing the presence of NAMPT in all three cellular compartments, whereas nicotinate phosphoribosyltransferase was predominantly cytosolic and mitochondrial, and nicotinamide riboside kinase mitochondrial and nuclear. In keeping with biosensor data, quinolinate phosphoribosyltransferase was expressed at extremely low levels. Innovation and Conclusions: Throughout this work, we validated the use of genetically encoded NAD+ biosensors to characterize subcellular distribution of NAD+ production routes in MM. The chance of real-time monitoring of NAD+ fluctuations after chemical perturbations, together with a deeper comprehension of the cofactor biosynthesis compartmentalization, strengthens the foundation for a targeted strategy of NAD+ pool manipulation in cancer and metabolic diseases.
Collapse
Affiliation(s)
- Federica Gaudino
- Department of Medical Sciences, University of Turin, Turin, Italy
| | | | - Antonella Managò
- Department of Medical Sciences, University of Turin, Turin, Italy
| | | | - Nadia Raffaelli
- Department of Clinical Sciences, Food and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
- Department of Agricultural, Food and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Tiziana Vaisitti
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Silvia Deaglio
- Department of Medical Sciences, University of Turin, Turin, Italy
| |
Collapse
|
21
|
Paulitschke V, Eichhoff O, Gerner C, Paulitschke P, Bileck A, Mohr T, Cheng PF, Leitner A, Guenova E, Saulite I, Freiberger SN, Irmisch A, Knapp B, Zila N, Chatziisaak T, Stephan J, Mangana J, Kunstfeld R, Pehamberger H, Aebersold R, Dummer R, Levesque MP. Proteomic identification of a marker signature for MAPKi resistance in melanoma. EMBO J 2019; 38:e95874. [PMID: 31267558 PMCID: PMC6669927 DOI: 10.15252/embj.201695874] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 05/15/2019] [Accepted: 05/22/2019] [Indexed: 12/21/2022] Open
Abstract
MAPK inhibitors (MAPKi) show outstanding clinical response rates in melanoma patients harbouring BRAF mutations, but resistance is common. The ability of melanoma cells to switch from melanocytic to mesenchymal phenotypes appears to be associated with therapeutic resistance. High-throughput, subcellular proteome analyses and RNAseq on two panels of primary melanoma cells that were either sensitive or resistant to MAPKi revealed that only 15 proteins were sufficient to distinguish between these phenotypes. The two proteins with the highest discriminatory power were PTRF and IGFBP7, which were both highly upregulated in the mesenchymal-resistant cells. Proteomic analysis of CRISPR/Cas-derived PTRF knockouts revealed targets involved in lysosomal activation, endocytosis, pH regulation, EMT, TGFβ signalling and cell migration and adhesion, as well as a significantly reduced invasive index and ability to form spheres in 3D culture. Overexpression of PTRF led to MAPKi resistance, increased cell adhesion and sphere formation. In addition, immunohistochemistry of patient samples showed that PTRF expression levels were a significant biomarker of poor progression-free survival, and IGFBP7 levels in patient sera were shown to be higher after relapse.
Collapse
Affiliation(s)
- Verena Paulitschke
- Department of DermatologyMedical University of ViennaViennaAustria
- Department of DermatologyUniversity of Zurich HospitalUniversity of ZurichZurichSwitzerland
- Department of BiologyInstitute of Molecular Systems BiologyETH ZurichZurichSwitzerland
| | - Ossia Eichhoff
- Department of DermatologyUniversity of Zurich HospitalUniversity of ZurichZurichSwitzerland
| | - Christopher Gerner
- Department of Analytical ChemistryFaculty of ChemistryUniversity of ViennaViennaAustria
| | - Philipp Paulitschke
- Institute of PhysicsCenter for NanoScienceLudwig Maximilians UniversityMunichGermany
| | - Andrea Bileck
- Department of Analytical ChemistryFaculty of ChemistryUniversity of ViennaViennaAustria
| | - Thomas Mohr
- Department of Medicine IInstitute of Cancer Research and Comprehensive Cancer CenterMedical University ViennaViennaAustria
| | - Phil F Cheng
- Department of DermatologyUniversity of Zurich HospitalUniversity of ZurichZurichSwitzerland
| | - Alexander Leitner
- Department of BiologyInstitute of Molecular Systems BiologyETH ZurichZurichSwitzerland
| | - Emmanuella Guenova
- Department of DermatologyUniversity of Zurich HospitalUniversity of ZurichZurichSwitzerland
| | - Ieva Saulite
- Department of DermatologyUniversity of Zurich HospitalUniversity of ZurichZurichSwitzerland
| | - Sandra N Freiberger
- Department of DermatologyUniversity of Zurich HospitalUniversity of ZurichZurichSwitzerland
| | - Anja Irmisch
- Department of DermatologyUniversity of Zurich HospitalUniversity of ZurichZurichSwitzerland
| | - Bernhard Knapp
- Department of StatisticsProtein Informatics GroupUniversity of OxfordOxfordUK
| | - Nina Zila
- Department of DermatologyMedical University of ViennaViennaAustria
| | | | - Jürgen Stephan
- Institute of PhysicsCenter for NanoScienceLudwig Maximilians UniversityMunichGermany
| | - Joanna Mangana
- Department of DermatologyUniversity of Zurich HospitalUniversity of ZurichZurichSwitzerland
| | - Rainer Kunstfeld
- Department of DermatologyMedical University of ViennaViennaAustria
| | | | - Ruedi Aebersold
- Department of BiologyInstitute of Molecular Systems BiologyETH ZurichZurichSwitzerland
- Faculty of ScienceUniversity of ZurichZurichSwitzerland
| | - Reinhard Dummer
- Department of DermatologyUniversity of Zurich HospitalUniversity of ZurichZurichSwitzerland
| | - Mitchell P Levesque
- Department of DermatologyUniversity of Zurich HospitalUniversity of ZurichZurichSwitzerland
| |
Collapse
|
22
|
Guo Q, Han N, Shi L, Yang L, Zhang X, Zhou Y, Yu S, Zhang M. NAMPT: A potential prognostic and therapeutic biomarker in patients with glioblastoma. Oncol Rep 2019; 42:963-972. [PMID: 31322259 PMCID: PMC6667917 DOI: 10.3892/or.2019.7227] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 07/03/2019] [Indexed: 12/19/2022] Open
Abstract
Glioblastoma (GBM) is the most common primary intracranial malignancy. GBM still exhibits high recurrence and mortality rates even following combined treatment with surgery, radiotherapy and chemotherapy, Therefore, the identification of novel therapeutic targets is urgent. Previous research has shown that nicotinamide phosphoribosyltransferase (NAMPT) plays a key role in cell metabolism and is closely related to the occurrence and development of many tumor types; yet, little is known concerning its relationship with GBM. Oncomine database analysis showed that the expression of NAMPT in GBM was higher than that in normal tissues; this finding was further confirmed by immunohistochemical staining of a tissue microarray. Data analysis with the R2 platform showed that patients with higher expression of NAMPT had worse prognoses than those with lower NAMPT expression. Using the GBM data in TCGA, four pathways enriched in the high NAMPT expression group were identified by gene set enrichment analysis (GSEA). NAMPT expression was knocked down in U87 and U251 GBM cells by lentiviral vectors carrying a small hairpin RNA (shRNA) targeting NAMPT. CCK-8, colony formation, wound healing, Transwell and apoptosis assays were carried out. The results showed that NAMPT knockdown decreased cell proliferation, migration, and invasion and promoted apoptosis. U87 GBM cells were used in a model of subcutaneous tumorigenesis in nude mice. The results showed that NAMPT knockdown slowed the growth of tumors in vivo. Therefore, we speculate that NAMPT may be a potential prognostic and therapeutic biomarker for glioblastoma.
Collapse
Affiliation(s)
- Qiuyun Guo
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Na Han
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Lei Shi
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Li Yang
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xiaoxi Zhang
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yangmei Zhou
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Shiying Yu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Mengxian Zhang
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
23
|
Audrito V, Managò A, Gaudino F, Deaglio S. Targeting metabolic reprogramming in metastatic melanoma: The key role of nicotinamide phosphoribosyltransferase (NAMPT). Semin Cell Dev Biol 2019; 98:192-201. [PMID: 31059816 DOI: 10.1016/j.semcdb.2019.05.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/02/2019] [Accepted: 05/02/2019] [Indexed: 12/13/2022]
Abstract
Cancer cells rewire their metabolism to support proliferation, growth and survival. In metastatic melanoma the BRAF oncogenic pathway is a master regulator of this process, highlighting the importance of metabolic reprogramming in the pathogenesis of this tumor and offering potential therapeutic approaches. Metabolic adaptation of melanoma cells generally requires increased amounts of NAD+, an essential redox cofactor in cellular metabolism and a signaling molecule. Nicotinamide phosphoribosyltransferase (NAMPT) is the most important NAD+ biosynthetic enzyme in mammalian cells and a direct target of the BRAF oncogenic signaling pathway. These findings suggest that NAMPT is an attractive new therapeutic target, particularly in combination strategies with BRAF or MEK inhibitors. Here we review current knowledge on how oncogenic signaling reprograms metabolism in BRAF-mutated melanoma, and discuss how NAMPT/NAD+ axis contributes to these processes. Lastly, we present evidence supporting a role of NAMPT as a novel therapeutic target in metastatic melanoma.
Collapse
Affiliation(s)
- Valentina Audrito
- Department of Medical Sciences, University of Turin, Turin, Italy; Italian Institute for Genomic Medicine, Turin, Italy.
| | - Antonella Managò
- Department of Medical Sciences, University of Turin, Turin, Italy; Italian Institute for Genomic Medicine, Turin, Italy
| | - Federica Gaudino
- Department of Medical Sciences, University of Turin, Turin, Italy; Italian Institute for Genomic Medicine, Turin, Italy
| | - Silvia Deaglio
- Department of Medical Sciences, University of Turin, Turin, Italy; Italian Institute for Genomic Medicine, Turin, Italy.
| |
Collapse
|
24
|
Lucena-Cacace A, Umeda M, Navas LE, Carnero A. NAMPT as a Dedifferentiation-Inducer Gene: NAD + as Core Axis for Glioma Cancer Stem-Like Cells Maintenance. Front Oncol 2019; 9:292. [PMID: 31119097 PMCID: PMC6507617 DOI: 10.3389/fonc.2019.00292] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 03/29/2019] [Indexed: 12/27/2022] Open
Abstract
Glioma Cancer Stem-Like Cells (GSCs) are a small subset of CD133+ cells with self-renewal properties and capable of initiating new tumors contributing to Glioma progression, maintenance, hierarchy, and complexity. GSCs are highly resistant to chemo and radiotherapy. These cells are believed to be responsible for tumor relapses and patients' fatal outcome after developing a recurrent Glioblastoma (GBM) or High Grade Glioma (HGG). GSCs are cells under replicative stress with high demands on NAD+ supply to repair DNA, maintain self-renewal capacity and to induce tumor plasticity. NAD+ feeds Poly-ADP polymerases (PARP) and NAD+-dependent deacetylases (SIRTUINS) contributing to GSC phenotype. This energetic core axis is mainly controlled by the rate-limiting enzyme nicotinamide phosphoribosyltransferase (NAMPT), an important oncogene contributing to tumor dedifferentiation. Targeting GSCs depicts a new frontier in Glioma therapy; hence NAMPT could represent a key regulator for GSCs maintenance. Its inhibition may attenuate GSCs properties by decreasing NAD+ supply, consequently contributing to a better outcome together with current therapies for Glioma control.
Collapse
Affiliation(s)
- Antonio Lucena-Cacace
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Masayuki Umeda
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Lola E Navas
- CIBERONC, ISCIII, Madrid, Spain.,Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío (HUVR), CSIC, Universidad de Sevilla, Sevilla, Spain
| | - Amancio Carnero
- CIBERONC, ISCIII, Madrid, Spain.,Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío (HUVR), CSIC, Universidad de Sevilla, Sevilla, Spain
| |
Collapse
|
25
|
Dalamaga M, Christodoulatos GS, Mantzoros CS. The role of extracellular and intracellular Nicotinamide phosphoribosyl-transferase in cancer: Diagnostic and therapeutic perspectives and challenges. Metabolism 2018; 82:72-87. [PMID: 29330025 DOI: 10.1016/j.metabol.2018.01.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 12/23/2017] [Accepted: 01/04/2018] [Indexed: 02/07/2023]
Abstract
Nicotinamide phosphoribosyl-transferase (Nampt) or pre-B cell colony-enhancing factor or visfatin represents a pleiotropic molecule acting as an enzyme, a cytokine and a growth factor. Intracellular Nampt plays an important role in cellular bioenergetics and metabolism, particularly NAD biosynthesis. NAD biosynthesis is critical in DNA repair, oncogenic signal transduction, transcription, genomic integrity and apoptosis. Although its insulin-mimetic function remains a controversial issue, extracellular Nampt presents proliferative, anti-apoptotic, pro-inflammatory, pro-angiogenic and metastatic properties. Nampt is upregulated in many malignancies, including obesity-associated cancers, and is associated with worse prognosis. Serum Nampt may be a potential diagnostic and prognostic biomarker in cancer. Pharmacologic agents that neutralize Nampt or medications that decrease Nampt levels or downregulate signaling pathways downstream of Nampt may prove to be useful anti-cancer treatments. In particular, Nampt inhibitors as monotherapy or in combination therapy have displayed anti-cancer activity in vivo and in vitro. The aim of this review is to explore the role of Nampt in cancer pathophysiology as well as to synopsize the mechanisms underlying the association between extracellular and intracellular Nampt, and malignancy. Exploring the interplay of cellular bioenergetics, inflammation and adiposopathy is expected to be of importance in the development of preventive and therapeutic strategies against cancer.
Collapse
Affiliation(s)
- Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Goudi, 11527 Athens, Greece.
| | - Gerasimos Socrates Christodoulatos
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Goudi, 11527 Athens, Greece; Department of Microbiology, KAT Hospital, Nikis 2, Kifisia, 14561 Athens, Greece
| | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; Section of Endocrinology, Boston VA Healthcare System, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
26
|
Ohanna M, Cerezo M, Nottet N, Bille K, Didier R, Beranger G, Mograbi B, Rocchi S, Yvan-Charvet L, Ballotti R, Bertolotto C. Pivotal role of NAMPT in the switch of melanoma cells toward an invasive and drug-resistant phenotype. Genes Dev 2018; 32:448-461. [PMID: 29567766 PMCID: PMC5900716 DOI: 10.1101/gad.305854.117] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 03/05/2018] [Indexed: 12/19/2022]
Abstract
In BRAFV600E melanoma cells, a global metabolomic analysis discloses a decrease in nicotinamide adenine dinucleotide (NAD+) levels upon PLX4032 treatment that is conveyed by a STAT5 inhibition and a transcriptional regulation of the nicotinamide phosphoribosyltransferase (NAMPT) gene. NAMPT inhibition decreases melanoma cell proliferation both in vitro and in vivo, while forced NAMPT expression renders melanoma cells resistant to PLX4032. NAMPT expression induces transcriptomic and epigenetic reshufflings that steer melanoma cells toward an invasive phenotype associated with resistance to targeted therapies and immunotherapies. Therefore, NAMPT, the key enzyme in the NAD+ salvage pathway, appears as a rational target in targeted therapy-resistant melanoma cells and a key player in phenotypic plasticity of melanoma cells.
Collapse
Affiliation(s)
- Mickaël Ohanna
- U1065, Institut National de la Santé et de la Recherche Médicale (INSERM), Biology and Pathologies of Melanocytes, Equipe Labellisée L'Association pour la Recherche sur le Cancer (ARC) 2015, Université Nice Côte d'Azur, INSERM, Centre Méditerranéen de Médecine Moléculaire (C3M), 06204 Nice, France
| | - Mickaël Cerezo
- U1065, Institut National de la Santé et de la Recherche Médicale (INSERM), Biology and Pathologies of Melanocytes, Equipe Labellisée L'Association pour la Recherche sur le Cancer (ARC) 2015, Université Nice Côte d'Azur, INSERM, Centre Méditerranéen de Médecine Moléculaire (C3M), 06204 Nice, France
| | - Nicolas Nottet
- Université Nice Côte d'Azur, INSERM, C3M, 06204 Nice, France
| | - Karine Bille
- U1065, Institut National de la Santé et de la Recherche Médicale (INSERM), Biology and Pathologies of Melanocytes, Equipe Labellisée L'Association pour la Recherche sur le Cancer (ARC) 2015, Université Nice Côte d'Azur, INSERM, Centre Méditerranéen de Médecine Moléculaire (C3M), 06204 Nice, France
| | - Robin Didier
- U1065, Institut National de la Santé et de la Recherche Médicale (INSERM), Biology and Pathologies of Melanocytes, Equipe Labellisée L'Association pour la Recherche sur le Cancer (ARC) 2015, Université Nice Côte d'Azur, INSERM, Centre Méditerranéen de Médecine Moléculaire (C3M), 06204 Nice, France
| | - Guillaume Beranger
- U1065, Institut National de la Santé et de la Recherche Médicale (INSERM), Biology and Pathologies of Melanocytes, Equipe Labellisée L'Association pour la Recherche sur le Cancer (ARC) 2015, Université Nice Côte d'Azur, INSERM, Centre Méditerranéen de Médecine Moléculaire (C3M), 06204 Nice, France
| | - Baharia Mograbi
- U1081, INSERM, Institute of Research on Cancer and Ageing of Nice (IRCAN), Equipe Labellisée ARC, Université Nice Côte d'Azur, UMR7284, Centre National de la Recherche Scientifique (CNRS), 06107 Nice, France
| | - Stéphane Rocchi
- U1065, Institut National de la Santé et de la Recherche Médicale (INSERM), Biology and Pathologies of Melanocytes, Equipe Labellisée L'Association pour la Recherche sur le Cancer (ARC) 2015, Université Nice Côte d'Azur, INSERM, Centre Méditerranéen de Médecine Moléculaire (C3M), 06204 Nice, France
| | - Laurent Yvan-Charvet
- U1065, INSERM, Team ATIP-Avenir, Université Nice Côte d'Azur, INSERM, C3M, 06204 Nice, France
| | - Robert Ballotti
- U1065, Institut National de la Santé et de la Recherche Médicale (INSERM), Biology and Pathologies of Melanocytes, Equipe Labellisée L'Association pour la Recherche sur le Cancer (ARC) 2015, Université Nice Côte d'Azur, INSERM, Centre Méditerranéen de Médecine Moléculaire (C3M), 06204 Nice, France
| | - Corine Bertolotto
- U1065, Institut National de la Santé et de la Recherche Médicale (INSERM), Biology and Pathologies of Melanocytes, Equipe Labellisée L'Association pour la Recherche sur le Cancer (ARC) 2015, Université Nice Côte d'Azur, INSERM, Centre Méditerranéen de Médecine Moléculaire (C3M), 06204 Nice, France
| |
Collapse
|
27
|
Zhou SJ, Bi TQ, Qin CX, Yang XQ, Pang K. Expression of NAMPT is associated with breast invasive ductal carcinoma development and prognosis. Oncol Lett 2018; 15:6648-6654. [PMID: 29725408 DOI: 10.3892/ol.2018.8164] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 01/19/2018] [Indexed: 01/29/2023] Open
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) possesses various functions in human cells, and altered NAMPT expression is associated with human carcinogenesis. The present study detected the expression of NAMPT in normal and cancerous breast tissues from 83 patients using immunohistochemistry, and analyzed its association with the clinicopathological and survival data of the patients. NAMPT was significantly overexpressed in the breast invasive ductal carcinoma tissues compared with adjacent normal mammary gland tissues. Upregulated NAMPT expression was associated with a larger tumor size, lymph node metastasis, advanced clinical tumor-node-metastasis stages, and estrogen receptor and progesterone receptor expression. Furthermore, NAMPT expression was associated with poor overall and disease-free survival in patients with breast cancer. In conclusion, NAMPT increased protein expression in tumor cells may contribute to the development and progression of breast invasive ductal carcinoma. Thus, detection of NAMPT expression might be useful as a biomarker for the early detection and prognosis prediction of breast cancer.
Collapse
Affiliation(s)
- Shao-Jie Zhou
- Department of Thyroid and Breast Surgery, Weihai Municipal Hospital, Weihai, Shandong 264200, P.R. China
| | - Tie-Qiang Bi
- Department of Thyroid and Breast Surgery, Weihai Municipal Hospital, Weihai, Shandong 264200, P.R. China
| | - Chun-Xin Qin
- Department of Thyroid and Breast Surgery, Weihai Municipal Hospital, Weihai, Shandong 264200, P.R. China
| | - Xiao-Qing Yang
- Department of Thyroid and Breast Surgery, Weihai Municipal Hospital, Weihai, Shandong 264200, P.R. China
| | - Kai Pang
- Department of Statistics, Weihai Municipal Hospital, Weihai, Shandong 264200, P.R. China
| |
Collapse
|
28
|
Audrito V, Managò A, La Vecchia S, Zamporlini F, Vitale N, Baroni G, Cignetto S, Serra S, Bologna C, Stingi A, Arruga F, Vaisitti T, Massi D, Mandalà M, Raffaelli N, Deaglio S. Nicotinamide Phosphoribosyltransferase (NAMPT) as a Therapeutic Target in BRAF-Mutated Metastatic Melanoma. J Natl Cancer Inst 2018; 110:290-303. [DOI: 10.1093/jnci/djx198] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Affiliation(s)
- Valentina Audrito
- Department of Medical Sciences, University of Turin, Italy
- Italian Institute for Genomic Medicine, Turin, Italy
| | - Antonella Managò
- Department of Medical Sciences, University of Turin, Italy
- Italian Institute for Genomic Medicine, Turin, Italy
| | - Sofia La Vecchia
- Department of Medical Sciences, University of Turin, Italy
- Italian Institute for Genomic Medicine, Turin, Italy
| | - Federica Zamporlini
- Department of Agricultural, Food and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Nicoletta Vitale
- Department of Molecular Biotechnologies and Health Science, University of Turin, Italy
| | - Gianna Baroni
- Division of Pathological Anatomy, Department of Surgery and Translational Medicine, University of Florence, Italy
| | - Simona Cignetto
- Department of Medical Sciences, University of Turin, Italy
- Italian Institute for Genomic Medicine, Turin, Italy
| | - Sara Serra
- Department of Medical Sciences, University of Turin, Italy
- Italian Institute for Genomic Medicine, Turin, Italy
| | - Cinzia Bologna
- Department of Medical Sciences, University of Turin, Italy
- Italian Institute for Genomic Medicine, Turin, Italy
| | - Aureliano Stingi
- Department of Medical Sciences, University of Turin, Italy
- Italian Institute for Genomic Medicine, Turin, Italy
| | - Francesca Arruga
- Department of Medical Sciences, University of Turin, Italy
- Italian Institute for Genomic Medicine, Turin, Italy
| | - Tiziana Vaisitti
- Department of Medical Sciences, University of Turin, Italy
- Italian Institute for Genomic Medicine, Turin, Italy
| | - Daniela Massi
- Division of Pathological Anatomy, Department of Surgery and Translational Medicine, University of Florence, Italy
| | - Mario Mandalà
- Unit of Medical Oncology, Department of Oncology and Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Nadia Raffaelli
- Department of Agricultural, Food and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Silvia Deaglio
- Department of Medical Sciences, University of Turin, Italy
- Italian Institute for Genomic Medicine, Turin, Italy
| |
Collapse
|
29
|
Buonvicino D, Mazzola F, Zamporlini F, Resta F, Ranieri G, Camaioni E, Muzzi M, Zecchi R, Pieraccini G, Dölle C, Calamante M, Bartolucci G, Ziegler M, Stecca B, Raffaelli N, Chiarugi A. Identification of the Nicotinamide Salvage Pathway as a New Toxification Route for Antimetabolites. Cell Chem Biol 2018; 25:471-482.e7. [PMID: 29478906 DOI: 10.1016/j.chembiol.2018.01.012] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 10/23/2017] [Accepted: 01/26/2018] [Indexed: 12/13/2022]
Abstract
Interest in the modulation of nicotinamide adenine dinucleotide (NAD) metabolome is gaining great momentum because of its therapeutic potential in different human disorders. Suppression of nicotinamide salvage by nicotinamide phosphoribosyl transferase (NAMPT) inhibitors, however, gave inconclusive results in neoplastic patients because several metabolic routes circumvent the enzymatic block converging directly on nicotinamide mononucleotide adenylyl transferases (NMNATs) for NAD synthesis. Unfortunately, NMNAT inhibitors have not been identified. Here, we report the identification of Vacor as a substrate metabolized by the consecutive action of NAMPT and NMNAT2 into the NAD analog Vacor adenine dinucleotide (VAD). This leads to inhibition of both enzymes, as well as NAD-dependent dehydrogenases, thereby causing unprecedented rapid NAD depletion, glycolytic block, energy failure, and necrotic death of NMNAT2-proficient cancer cells. Conversely, lack of NMNAT2 expression confers complete resistance to Vacor. Remarkably, Vacor prompts VAD formation and growth suppression in NMNAT2-positive neuroblastoma and melanoma xenografts. Our data show the first evidence of harnessing the entire nicotinamide salvage pathway for antimetabolic strategies.
Collapse
Affiliation(s)
- Daniela Buonvicino
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence 50139, Italy
| | - Francesca Mazzola
- Department of Clinical Science, Polytechnic University of Marche, Ancona 60131, Italy
| | - Federica Zamporlini
- Department of Agricultural, Food and Environmental Sciences, Polytechnic University of Marche, Ancona 60131, Italy
| | - Francesco Resta
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence 50139, Italy
| | - Giuseppe Ranieri
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence 50139, Italy
| | - Emidio Camaioni
- Department of Pharmaceutical Sciences, University of Perugia, Perugia 06123, Italy
| | - Mirko Muzzi
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence 50139, Italy
| | - Riccardo Zecchi
- Mass Spectrometry Service Centre (CISM), University of Florence, Florence 50139, Italy
| | - Giuseppe Pieraccini
- Mass Spectrometry Service Centre (CISM), University of Florence, Florence 50139, Italy
| | - Christian Dölle
- Department of Molecular Biology, University of Bergen, 5020 Bergen, Norway
| | - Massimo Calamante
- Department of Chemistry, University of Florence, Florence 50019, Italy
| | - Gianluca Bartolucci
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence 50139, Italy
| | - Mathias Ziegler
- Department of Molecular Biology, University of Bergen, 5020 Bergen, Norway
| | - Barbara Stecca
- Core Research Laboratory-Istituto Toscano Tumori, Department of Oncology, Careggi University Hospital, Florence 50139, Italy
| | - Nadia Raffaelli
- Department of Agricultural, Food and Environmental Sciences, Polytechnic University of Marche, Ancona 60131, Italy
| | - Alberto Chiarugi
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence 50139, Italy.
| |
Collapse
|
30
|
Palacios DS, Meredith E, Kawanami T, Adams C, Chen X, Darsigny V, Geno E, Palermo M, Baird D, Boynton G, Busby SA, George EL, Guy C, Hewett J, Tierney L, Thigale S, Weihofen W, Wang L, White N, Yin M, Argikar UA. Structure based design of nicotinamide phosphoribosyltransferase (NAMPT) inhibitors from a phenotypic screen. Bioorg Med Chem Lett 2018; 28:365-370. [DOI: 10.1016/j.bmcl.2017.12.037] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 12/13/2017] [Accepted: 12/16/2017] [Indexed: 10/18/2022]
|
31
|
Vora M, Alattia LA, Ansari J, Ong M, Cotelingam J, Coppola D, Shackelford R. Nicotinamide Phosphoribosyl Transferase a Reliable Marker of Progression in Cervical Dysplasia. Anticancer Res 2017; 37:4821-4825. [PMID: 28870901 DOI: 10.21873/anticanres.11889] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 06/29/2017] [Accepted: 07/12/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND/AIM Nicotinamide phosphoribosyl transferase (Nampt) catalyses the rate-limiting step of the mammalian nicotinamide adenine dinucleotide (NAD) salvage pathway. Nampt is highly expressed in several epithelial and mesenchymal neoplasms, where is promotes cell-cycle progression ans chemotherapy resistance. To our knowledge, alterations in Nampt expression have not been examined in cervical intraepithelial neoplasia (CIN) or squamous cell carcinoma (SCC). MATERIALS AND METHODS We performed immunohistochemical analysis for Nampt using tissue microarrays on 14 samples of benign cervical squamous epithelium and 15 CIN I, 15 CIN II, and 13 samples of CIN III. The SCCs included 5 low-grade, 67 intermediate-grade, and 81 high-grade tumors. RESULTS Nampt levels increased with increased CIN grades were compared to benign cervical squamous epithelium. Similarly, Nampt levels increased with increasing SCC grade. CONCLUSION Nampt expression is a reliable marker of progression in cervical dysplasia and SCC.
Collapse
Affiliation(s)
- Moiz Vora
- Department of Pathology and Translational Pathobiology, LSU Health, Shreveport, LA, U.S.A
| | - Lubna A Alattia
- Department of Pathology and Translational Pathobiology, LSU Health, Shreveport, LA, U.S.A
| | - Junaid Ansari
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center, Shreveport, LA, U.S.A
| | - Menchu Ong
- Department of Pathology and Translational Pathobiology, LSU Health, Shreveport, LA, U.S.A
| | - James Cotelingam
- Department of Pathology and Translational Pathobiology, LSU Health, Shreveport, LA, U.S.A
| | - Domenico Coppola
- Department of Anatomic Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, U.S.A
| | - Rodney Shackelford
- Department of Pathology and Translational Pathobiology, LSU Health, Shreveport, LA, U.S.A.
| |
Collapse
|
32
|
The NAMPT/E2F2/SIRT1 axis promotes proliferation and inhibits p53-dependent apoptosis in human melanoma cells. Biochem Biophys Res Commun 2017; 493:77-84. [PMID: 28919418 DOI: 10.1016/j.bbrc.2017.09.071] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 09/13/2017] [Indexed: 02/08/2023]
Abstract
Melanoma is the most common primary malignant neoplasm in adults, causing more deaths than any other skin cancer, necessitating the development of new target-based approaches. Current evidence suggests SIRT1, the mammalian nicotinamide adenine dinucleotide (NAD+)-dependent protein deacetylase, and nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting NAD+ biosynthetic enzyme, together comprise a novel systemic regulatory network to play a pivotal role in cell proliferation and apoptosis. Nevertheless, how the regulation of this cofactor interfaces with signal transduction network remains poorly understood in melanoma. Here, we report NAMPT is highly expressed in melanomaassociated with poor overall survival in patients. Pharmacological and genetic inhibition of NAMPT decreased NAD+ levels and melanoma cell proliferation capacity, and NAMPT knockdown induced apoptosis through the activity of the tumor suppressor p53. Next, we demonstrate NAMPT regulates the transcription factor E2F family member 2 (E2F2) in the apoptosis process. Downstream, E2F2 control the mRNA and protein levels of SIRT1. Finally, we find NAMPT mediates the apoptosis resistance of melanoma cells through NAMPT-E2F2-SIRT1 axis, more than NAD+-driven transcriptional program. Accordingly, our results demonstrated that NAMPT is a prognostic marker in melanoma, and the identificationofNAMPT-E2F2-SIRT1 pathway establishes another link between NAMPT and apoptosis events in melanoma, with therapeutic implications for this deadly cancer.
Collapse
|
33
|
Synthesis and Degradation of Adenosine 5'-Tetraphosphate by Nicotinamide and Nicotinate Phosphoribosyltransferases. Cell Chem Biol 2017; 24:553-564.e4. [PMID: 28416276 DOI: 10.1016/j.chembiol.2017.03.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 02/03/2017] [Accepted: 03/14/2017] [Indexed: 12/24/2022]
Abstract
Adenosine 5'-tetraphosphate (Ap4) is a ubiquitous metabolite involved in cell signaling in mammals. Its full physiological significance remains unknown. Here we show that two enzymes committed to NAD biosynthesis, nicotinamide phosphoribosyltransferase (NAMPT) and nicotinate phosphoribosyltransferase (NAPT), can both catalyze the synthesis and degradation of Ap4 through their facultative ATPase activity. We propose a mechanism for this unforeseen additional reaction, and demonstrate its evolutionary conservation in bacterial orthologs of mammalian NAMPT and NAPT. Furthermore, evolutionary distant forms of NAMPT were inhibited in vitro by the FK866 drug but, remarkably, it does not block synthesis of Ap4. In fact, FK866-treated murine cells showed decreased NAD but increased Ap4 levels. Finally, murine cells and plasma with engineered or naturally fluctuating NAMPT levels showed matching Ap4 fluctuations. These results suggest a role of Ap4 in the actions of NAMPT, and prompt to evaluate the role of Ap4 production in the actions of NAMPT inhibitors.
Collapse
|
34
|
Kennedy BE, Sharif T, Martell E, Dai C, Kim Y, Lee PWK, Gujar SA. NAD + salvage pathway in cancer metabolism and therapy. Pharmacol Res 2016; 114:274-283. [PMID: 27816507 DOI: 10.1016/j.phrs.2016.10.027] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 10/30/2016] [Indexed: 12/22/2022]
Abstract
Nicotinamide adenine dinucleotide (NAD+) is an essential coenzyme for various physiological processes including energy metabolism, DNA repair, cell growth, and cell death. Many of these pathways are typically dysregulated in cancer cells, making NAD+ an intriguing target for cancer therapeutics. NAD+ is mainly synthesized by the NAD+ salvage pathway in cancer cells, and not surprisingly, the pharmacological targeting of the NAD+ salvage pathway causes cancer cell cytotoxicity in vitro and in vivo. Several studies have described the precise consequences of NAD+ depletion on cancer biology, and have demonstrated that NAD+ depletion results in depletion of energy levels through lowered rates of glycolysis, reduced citric acid cycle activity, and decreased oxidative phosphorylation. Additionally, depletion of NAD+ causes sensitization of cancer cells to oxidative damage by disruption of the anti-oxidant defense system, decreased cell proliferation, and initiation of cell death through manipulation of cell signaling pathways (e.g., SIRT1 and p53). Recently, studies have explored the effect of well-known cancer therapeutics in combination with pharmacological depletion of NAD+ levels, and found in many cases a synergistic effect on cancer cell cytotoxicity. In this context, we will discuss the effects of NAD+ salvage pathway inhibition on cancer cell biology and provide insight on this pathway as a novel anti-cancer therapeutic target.
Collapse
Affiliation(s)
- Barry E Kennedy
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada
| | - Tanveer Sharif
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada
| | - Emma Martell
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada
| | - Cathleen Dai
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada
| | - Youra Kim
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| | - Patrick W K Lee
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada; Department of Pathology, Dalhousie University, Halifax, NS, Canada
| | - Shashi A Gujar
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada; Department of Pathology, Dalhousie University, Halifax, NS, Canada; Centre for Innovative and Collaborative Health Systems Research, IWK Health Centre, Halifax, NS, Canada.
| |
Collapse
|
35
|
Wang G, Tian W, Liu Y, Ju Y, Shen Y, Zhao S, Zhang B, Li Y. Visfatin Triggers the Cell Motility of Non-Small Cell Lung Cancer via Up-Regulation of Matrix Metalloproteinases. Basic Clin Pharmacol Toxicol 2016; 119:548-554. [PMID: 27224551 DOI: 10.1111/bcpt.12623] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 05/09/2016] [Indexed: 12/21/2022]
Abstract
High levels of visfatin are correlated with worse clinical prognosis of various cancers. Still, the effects and mechanisms of visfatin on progression of non-small cell lung cancer (NSCLC) remain unclear. This study revealed that plasma levels of visfatin in patients with NSCLC (585 ± 287 pg/ml) were significantly (p < 0.01) higher than those in healthy people (142 ± 61.1 pg/ml). The high level of plasma visfatin was found to be significantly (p < 0.05) correlated with TNM stage, lymph node metastasis and distant metastasis. Visfatin treatment can increase the migration and invasion of NSCLC cells via up-regulation of metalloproteinase-2 (MMP-2) and MMP-9. Both si-MMP-2 and si-MMP-9 attenuated visfatin-induced migration of NSCLC cells. The inhibitor of NF-κB, while not ERK1/2, p38-MAPK or PI3K/Akt, can significantly abolish visfatin-induced migration of A549 cells and up-regulation of MMP-2 and MMP-9. Furthermore, visfatin can increase the phosphorylation of IκBα and p65 and the transcription activities of NF-κB in NSCLC cells. ACHP, the inhibitor of IKK-β, blocked visfatin-induced activation of p65 and up-regulation of MMP-2 and MMP-9. Collectively, our data revealed that visfatin can trigger the in vitro migration and invasion of NSCLC cells via up-regulation of MMPs through activation of NF-κB.
Collapse
Affiliation(s)
- Guanghai Wang
- Department of Respiratory Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, China
| | - Wenjun Tian
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Yiqing Liu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Ying Ju
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Yajuan Shen
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Shengmei Zhao
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Bingchang Zhang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Yu Li
- Department of Respiratory Medicine, Qilu Hospital, Shandong University, Jinan, Shandong, China
| |
Collapse
|
36
|
Chen H, Wang S, Zhang H, Nice EC, Huang C. Nicotinamide phosphoribosyltransferase (Nampt) in carcinogenesis: new clinical opportunities. Expert Rev Anticancer Ther 2016; 16:827-38. [PMID: 27186719 DOI: 10.1080/14737140.2016.1190649] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Nicotinamide phosphoribosyltransferase (Nampt) is the rate-limiting enzyme that catalyzes the first step in the mammalian nicotinamide adenine dinucleotide (NAD) salvage pathway. Aberrant NAD metabolism was associated with oncogenic signal transduction, suggesting the critical roles of Nampt in tumorigenesis and metastasis. Additionally, Nampt can be secreted out of the cell, and this extracellular form of Nampt (eNampt) was shown to induce inflammation and angiogenesis due to its cytokine activity, which may also be involved in carcinogenesis. AREAS COVERED This article reviews recent advances in the studies of Nampt in carcinogenesis, with a special highlight on Nampt inhibitors and future clinical application, including cancer diagnosis, prognosis and therapy. Expert commentary: Nampt not only maintains the balance of cellular metabolism, but also has a profound influence on multiple aspects of carcinogenesis. Therefore, elucidation of these mechanisms opens the door for future clinical applications targeting this protein. Additional studies are needed to address important questions including the relationship between extracellular Nampt and carcinogenesis.
Collapse
Affiliation(s)
- Hang Chen
- a Key Laboratory of Tropical Diseases and Translational Medicine of Ministry of Education & Department of Neurology , The Affiliated Hospital of Hainan Medical College , Haikou , China
| | - Shiyu Wang
- a Key Laboratory of Tropical Diseases and Translational Medicine of Ministry of Education & Department of Neurology , The Affiliated Hospital of Hainan Medical College , Haikou , China
| | - Haiyuan Zhang
- a Key Laboratory of Tropical Diseases and Translational Medicine of Ministry of Education & Department of Neurology , The Affiliated Hospital of Hainan Medical College , Haikou , China
| | - Edouard C Nice
- b Department of Biochemistry and Molecular Biology , Monash University , Clayton , Australia
| | - Canhua Huang
- c State Key Laboratory for Biotherapy and Cancer Center, West China Hospital , Sichuan University, and Collaborative Innovation Center of Biotherapy , Chengdu , China
| |
Collapse
|
37
|
Yang J, Zhang K, Song H, Wu M, Li J, Yong Z, Jiang S, Kuang X, Zhang T. Visfatin is involved in promotion of colorectal carcinoma malignancy through an inducing EMT mechanism. Oncotarget 2016; 7:32306-17. [PMID: 27058759 PMCID: PMC5078014 DOI: 10.18632/oncotarget.8615] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 03/28/2016] [Indexed: 02/05/2023] Open
Abstract
Increasing evidences suggested visfatin, a newly discovered obesity-induced adipocytokine, is involved in promotion of cancer malignancy and correlated with worse clinical prognosis. While its effects and mechanisms on progression of colorectal cancer (CRC) remain unclear. Our clinical data show that visfatin protein is over expressed, positive associated with lymph node metastasis, high-grade tumor, and poor prognosis in 87 CRC patients. The levels of plasma visfatin are significantly upregulated in Stage IV colon cancer. Visfatin can significantly promote the in vitro migration and invasion of CRC cells via induction epithelial mesenchymal transition (EMT). It can increase the expression and nuclear translocation of Snail, a key transcription factor in regulating EMT. While silencing of Snail attenuates visfatin induced EMT. Further studies reveal visfatin can inhibit the association of Snail with GSK-3β and subsequently suppress ubiquitylation of Snail. In addition, visfatin can increase the expression and nuclear translocation of β-catenin, elevate its binding with Snail promoter, and then increase the transcription of Snail. While inhibitor of PI3K/Akt, LY294002, abolishes visfatin induced up regulation of Snail, Vimentin (Vim), β-catenin, and phosphorylated GSK-3β. In summary, our data suggest that increased expression of visfatin are associated with a more aggressive phenotype of CRC patients. It can trigger the EMT of CRC cells via Akt/GSK-3β/β-catenin signals.
Collapse
Affiliation(s)
- Jing Yang
- School of Biomedical Sciences, Chengdu Medical College, Chengdu, China
| | - Kun Zhang
- School of Biomedical Sciences, Chengdu Medical College, Chengdu, China
| | - Haixing Song
- School of Biomedical Sciences, Chengdu Medical College, Chengdu, China
| | - Mingbo Wu
- School of Biomedical Sciences, Chengdu Medical College, Chengdu, China
| | - Jingyi Li
- School of Biomedical Sciences, Chengdu Medical College, Chengdu, China
| | - Ziyi Yong
- School of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Sheng Jiang
- School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
| | - Xi Kuang
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Sichuan, China
| | - Tao Zhang
- School of Biomedical Sciences, Chengdu Medical College, Chengdu, China
| |
Collapse
|
38
|
ZHU YANYAN, GUO MEIYAN, ZHANG LINGYUN, XU TAO, WANG LI, XU GUOXIONG. Biomarker triplet NAMPT/VEGF/HER2 as a de novo detection panel for the diagnosis and prognosis of human breast cancer. Oncol Rep 2016; 35:454-462. [DOI: 10.3892/or.2015.4391] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 09/23/2015] [Indexed: 11/05/2022] Open
|
39
|
Grolla AA, Torretta S, Gnemmi I, Amoruso A, Orsomando G, Gatti M, Caldarelli A, Lim D, Penengo L, Brunelleschi S, Genazzani AA, Travelli C. Nicotinamide phosphoribosyltransferase (NAMPT/PBEF/visfatin) is a tumoural cytokine released from melanoma. Pigment Cell Melanoma Res 2015; 28:718-29. [DOI: 10.1111/pcmr.12420] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 09/08/2015] [Indexed: 12/20/2022]
Affiliation(s)
- Ambra A. Grolla
- Department of Pharmaceutical Sciences and Department of Medical Sciences; Università del Piemonte Orientale; Novara Italy
| | - Simone Torretta
- Department of Pharmaceutical Sciences and Department of Medical Sciences; Università del Piemonte Orientale; Novara Italy
| | - Ilaria Gnemmi
- Department of Pharmaceutical Sciences and Department of Medical Sciences; Università del Piemonte Orientale; Novara Italy
| | - Angela Amoruso
- Department of Pharmaceutical Sciences and Department of Medical Sciences; Università del Piemonte Orientale; Novara Italy
| | - Giuseppe Orsomando
- Section of Biochemistry; Department of Clinical Sciences; Polytechnic University of Marche; Ancona Italy
| | - Marco Gatti
- Department of Pharmaceutical Sciences and Department of Medical Sciences; Università del Piemonte Orientale; Novara Italy
| | - Antonio Caldarelli
- Department of Pharmaceutical Sciences and Department of Medical Sciences; Università del Piemonte Orientale; Novara Italy
| | - Dmitry Lim
- Department of Pharmaceutical Sciences and Department of Medical Sciences; Università del Piemonte Orientale; Novara Italy
| | - Lorenza Penengo
- Department of Pharmaceutical Sciences and Department of Medical Sciences; Università del Piemonte Orientale; Novara Italy
- Institute of Pharmacology and Toxicology; University of Zürich-Vetsuisse; Zürich Switzerland
| | - Sandra Brunelleschi
- Department of Pharmaceutical Sciences and Department of Medical Sciences; Università del Piemonte Orientale; Novara Italy
| | - Armando A. Genazzani
- Department of Pharmaceutical Sciences and Department of Medical Sciences; Università del Piemonte Orientale; Novara Italy
| | - Cristina Travelli
- Department of Pharmaceutical Sciences and Department of Medical Sciences; Università del Piemonte Orientale; Novara Italy
| |
Collapse
|
40
|
Lv X, Zhang L, Zhu Y, Said HM, Shi J, Xu G. Regulative Effect of Nampt on Tumor Progression and Cell Viability in Human Colorectal Cancer. J Cancer 2015; 6:849-858. [PMID: 26284136 PMCID: PMC4532982 DOI: 10.7150/jca.12341] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 06/16/2015] [Indexed: 01/07/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer disease. Here we examined Nampt expression in patients with CRC and the effect of Nampt on cell viability in CRC cells. Nampt protein was overexpressed in colorectal adenoma as well as colorectal carcinoma. The immunoreactive staining of Nampt was negative in the adjacent normal colorectal tissue, weak in colorectal adenoma, and strong in colorectal carcinoma, which may represent tumor progression. Further evaluation of clinical data showed that Nampt expression was not correlated with the clinicopathological characteristics of CRC. Additionally, our in vitro studies demonstrated that Nampt promotes CRC cell viability, whereas the Nampt inhibitor FK866 suppressed CRC cell viability, which was in concordance with the previous studies in other cancer cells. Treatment with Nampt-siRNA reduced the Nampt protein expression resulting in the inhibition of the cell viability of HCT116 and Caco2. Thus, the involvement of Nampt in cell growth indicates that Nampt may play an important role in colorectal tumorigenesis. As a consequence, our results suggest that Nampt may be considered as a progression marker of colorectal tumor and a potentially therapeutic target for the treatment of CRC.
Collapse
Affiliation(s)
- Xiaoqun Lv
- 1. Department of Pharmacy, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - Lingyun Zhang
- 2. Center Laboratory, Jinshan Hospital, Fudan University, Shanghai 201508, China
- 3. Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yanyan Zhu
- 4. Department of General Surgery, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - Harun M. Said
- 5. Department of Radiation Oncology, University of Würzburg, D-97080 Würzburg, Germany
| | - Jimin Shi
- 2. Center Laboratory, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - Guoxiong Xu
- 2. Center Laboratory, Jinshan Hospital, Fudan University, Shanghai 201508, China
- 3. Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
41
|
Sampath D, Zabka TS, Misner DL, O’Brien T, Dragovich PS. Inhibition of nicotinamide phosphoribosyltransferase (NAMPT) as a therapeutic strategy in cancer. Pharmacol Ther 2015; 151:16-31. [DOI: 10.1016/j.pharmthera.2015.02.004] [Citation(s) in RCA: 162] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 02/02/2015] [Indexed: 12/12/2022]
|
42
|
Roulston A, Shore GC. New strategies to maximize therapeutic opportunities for NAMPT inhibitors in oncology. Mol Cell Oncol 2015; 3:e1052180. [PMID: 27308565 PMCID: PMC4845202 DOI: 10.1080/23723556.2015.1052180] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 05/13/2015] [Indexed: 12/16/2022]
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) is crucial for nicotinamide adenine dinucleotide (NAD(+)) biosynthesis in mammalian cells. NAMPT inhibitors represent multifunctional anticancer agents that act on NAD(+) metabolism to shut down glycolysis, nucleotide biosynthesis, and ATP generation and act indirectly as PARP and sirtuin inhibitors. The selectivity of NAMPT inhibitors preys on the increased metabolic requirements to replenish NAD(+) in cancer cells. Although initial clinical studies with NAMPT inhibitors did not achieve single-agent therapeutic levels before dose-limiting toxicities were reached, a new understanding of alternative rescue pathways and a biomarker that can be used to select patients provides new opportunities to widen the therapeutic window and achieve efficacious doses in the clinic. Recent work has also illustrated the potential for drug combination strategies to further enhance the therapeutic opportunities. This review summarizes recent discoveries in NAD(+)/NAMPT inhibitor biology in the context of exploiting this new knowledge to optimize the clinical outcomes for this promising new class of agents.
Collapse
Affiliation(s)
- Anne Roulston
- Laboratory for Therapeutic Development, Rosalind and Morris Goodman Cancer Research Centre, and Dept. Biochemistry, McGill University , Montreal, QC, Canada
| | - Gordon C Shore
- Laboratory for Therapeutic Development, Rosalind and Morris Goodman Cancer Research Centre, and Dept. Biochemistry, McGill University , Montreal, QC, Canada
| |
Collapse
|
43
|
Lu GW, Wang QJ, Xia MM, Qian J. Elevated plasma visfatin levels correlate with poor prognosis of gastric cancer patients. Peptides 2014; 58:60-4. [PMID: 24911837 DOI: 10.1016/j.peptides.2014.05.016] [Citation(s) in RCA: 24] [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: 04/26/2014] [Revised: 05/30/2014] [Accepted: 05/30/2014] [Indexed: 01/29/2023]
Abstract
Visfatin is a proinflammmatory cytokine with accumulating evidence for its rise in circulation of cancer patients. This study aimed to evaluate the relationship between preoperative plasma visfatin level and prognosis of gastric cancers. Preoperative plasma visfatin levels of 262 patients with gastric cancers and plasma visfatin levels of 262 healthy individuals were determined using enzyme-linked immunosorbent assay. Preoperative plasma visfatin level was substantially higher in patients than in healthy subjects. Plasma visfatin levels were associated with invasion depth, lymph node metastasis, distant metastasis, peritoneal dissemination, tumor size and tumor node metastasis stage. Multivariate analysis revealed that high plasma visfatin level was an independent factor for death. Receiver operating characteristic curve analysis showed that plasma visfatin level predicted death with high area under curve. Multivariate Cox regression analysis identified plasma visfatin level as an independent predictor of overall survival. Thus, our results suggest that high preoperative plasma visfatin level is associated with prognostic factors for gastric cancer as well as may play a role as prognostic biomarker in gastric cancer survival.
Collapse
Affiliation(s)
- Guo-Wen Lu
- Department of Oncological Surgery, Yinzhou People's Hospital, Yinzhou Hospital Affiliated to Medical School of Ningbo University, 251 Baizhang East Road, Ningbo 315040, China
| | - Qi-Jun Wang
- Department of Clinical Laboratory, Yinzhou People's Hospital, Yinzhou Hospital Affiliated to Medical School of Ningbo University, 251 Baizhang East Road, Ningbo 315040, China
| | - Min-Ming Xia
- Department of Oncological Surgery, Yinzhou People's Hospital, Yinzhou Hospital Affiliated to Medical School of Ningbo University, 251 Baizhang East Road, Ningbo 315040, China
| | - Jiao Qian
- Department of Oncological Surgery, Yinzhou People's Hospital, Yinzhou Hospital Affiliated to Medical School of Ningbo University, 251 Baizhang East Road, Ningbo 315040, China.
| |
Collapse
|
44
|
Shackelford RE, Mayhall K, Maxwell NM, Kandil E, Coppola D. Nicotinamide phosphoribosyltransferase in malignancy: a review. Genes Cancer 2014; 4:447-56. [PMID: 24386506 DOI: 10.1177/1947601913507576] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 08/26/2013] [Indexed: 12/15/2022] Open
Abstract
Nicotinamide phosphoribosyltransferase (Nampt) catalyzes the rate-limiting step of nicotinamide adenine dinucleotide (NAD) synthesis. Both intracellular and extracellular Nampt (iNampt and eNampt) levels are increased in several human malignancies and some studies demonstrate increased iNampt in more aggressive/invasive tumors and in tumor metastases. Several different molecular targets have been identified that promote carcinogenesis following iNampt overexpression, including SirT1, CtBP, and PARP-1. Additionally, eNampt is elevated in several human cancers and is often associated with a higher tumor stage and worse prognoses. Here we review the roles of Nampt in malignancy, some of the known mechanisms by which it promotes carcinogenesis, and discuss the possibility of employing Nampt inhibitors in cancer treatment.
Collapse
Affiliation(s)
| | - Kim Mayhall
- Tulane University School of Medicine, New Orleans, LA, USA
| | | | - Emad Kandil
- Tulane University School of Medicine, New Orleans, LA, USA
| | - Domenico Coppola
- Anatomic Pathology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| |
Collapse
|
45
|
Visfatin concentrations in children with leukemia before and after stem cell transplantation. Exp Hematol 2014; 42:252-60. [DOI: 10.1016/j.exphem.2013.12.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 11/16/2013] [Accepted: 12/16/2013] [Indexed: 11/23/2022]
|
46
|
Enninga EAL, Holtan SG, Creedon DJ, Dronca RS, Nevala WK, Ognjanovic S, Markovic SN. Immunomodulatory effects of sex hormones: requirements for pregnancy and relevance in melanoma. Mayo Clin Proc 2014; 89:520-35. [PMID: 24684874 PMCID: PMC4286150 DOI: 10.1016/j.mayocp.2014.01.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Revised: 12/18/2013] [Accepted: 01/02/2014] [Indexed: 12/20/2022]
Abstract
Similarities between the pathologic progression of cancer and the physiologic process of placentation (eg, proliferation, invasion, and local/systemic tolerance) have been recognized for many years. Sex hormones such as human chorionic gonadotropin, estrogens, progesterone, and others contribute to induction of immunologic tolerance at the beginning of gestation. Sex hormones have been shown to play contributory roles in the growth of cancers such as breast cancer, prostrate cancer, endometrial cancer, and ovarian cancer, but their involvement as putative mediators of the immunologic escape of cancer is still being elucidated. Herein, we compare the emerging mechanism by which sex hormones modulate systemic immunity in pregnancy and their potentially similar role in cancer. To do this, we conducted a PubMed search using combinations of the following keywords: "immune regulation," "sex hormones," "pregnancy," "melanoma," and "cancer." We did not limit our search to specific publication dates. Mimicking the maternal immune response to pregnancy, especially in late gestation, might aid in design of better therapies to reconstitute endogenous antitumor immunity and improve survival.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Svetomir N Markovic
- Department of Oncology, Mayo Clinic, Rochester, MN; Department of Medicine, Division of Hematology, Mayo Clinic, Rochester, MN.
| |
Collapse
|
47
|
Galli U, Travelli C, Massarotti A, Fakhfouri G, Rahimian R, Tron GC, Genazzani AA. Medicinal chemistry of nicotinamide phosphoribosyltransferase (NAMPT) inhibitors. J Med Chem 2013; 56:6279-96. [PMID: 23679915 DOI: 10.1021/jm4001049] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Nicotinamide phoshophoribosyltransferase (NAMPT) plays a key role in the replenishment of the NAD pool in cells. This in turn makes this enzyme an important player in bioenergetics and in the regulation of NAD-using enzymes, such as PARPs and sirtuins. Furthermore, there is now ample evidence that NAMPT is secreted and has a role as a cytokine. An important role of either the intracellular or extracellular form of NAMPT has been shown in cancer, inflammation, and metabolic diseases. The first NAMPT inhibitors (FK866 and CHS828) have already entered clinical trials, and a surge in interest in the synthesis of novel molecules has occurred. The present review summarizes the recent progress in this field.
Collapse
Affiliation(s)
- Ubaldina Galli
- Dipartimento di Scienze del Farmaco, Università degli Studi del Piemonte Orientale "A. Avogadro", Largo Donegani 2, 28100 Novara, Italy
| | | | | | | | | | | | | |
Collapse
|