1
|
Meng X, Cao J, Zheng H, Ma X, Wang Y, Tong Y, Xie S, Lu R, Guo L. TPX2 promotes ovarian tumorigenesis by interacting with Lamin A/C and affecting its stability. Cancer Med 2023; 12:9738-9748. [PMID: 36789877 PMCID: PMC10166900 DOI: 10.1002/cam4.5683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/31/2023] [Indexed: 02/16/2023] Open
Abstract
OBJECTIVE Ovarian cancer (OC) is one of the fatal gynecologic malignancies. However, there are no effective prognostic or therapeutic indicators for OC. Herein, we aim to reveal the potential function of targeting protein for Xklp2 (TPX2) in OC progression. METHODS Immunohistochemical and bioinformatic analyses were used to evaluate the level of TPX2 in OC samples. Effects of TPX2 on cell proliferation, cell apoptosis and ROS production were evaluated in vivo and in vitro. Mass spectrometry, Co-IP and immunofluorescence assays were performed to identify and verify protein-protein interactions. RESULTS Our data showed that pathological overexpression (OE) of the TPX2 in OC could manifest a poor prognosis. Functional studies demonstrated that TPX2 silencing led to the suppression of cell proliferation in vitro and in vivo through an increase in reactive oxygen species (ROS) level and apoptosis, while TPX2 OE exhibited the opposite effect. Furthermore, by mass spectrometric analysis, we identified a novel interacting partner, Lamin A/C, for TPX2. Mechanistically, TPX2 regulated Lamin A/C's stability by modulating the level of phospho-Lamin A/C (Ser 22). CONCLUSION Our findings thus suggest that TPX2 may be a promising therapeutic target for OC.
Collapse
Affiliation(s)
- Xin Meng
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jiazhen Cao
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hui Zheng
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xiaolu Ma
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yanchun Wang
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Ying Tong
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Suhong Xie
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Renquan Lu
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lin Guo
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
2
|
The Kynurenine Pathway and Cancer: Why Keep It Simple When You Can Make It Complicated. Cancers (Basel) 2022; 14:cancers14112793. [PMID: 35681770 PMCID: PMC9179486 DOI: 10.3390/cancers14112793] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/01/2022] [Accepted: 06/03/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary The kynurenine pathway has two main physiological roles: (i) it protects specific organs such as the eyes and placenta from strong immune reactions and (ii) it additionally generate in the liver and kidney a metabolite essential to all cells of human body. Abnormal activation of this pathway is recurrently observed in numerous cancer types. Its two functions are hijacked to promote tumor growth and cancer cell dissemination through multiple mechanisms. Clinical assays including administration of inhibitors of this pathway have not yet been successful. The complex regulation of this pathway is likely the reason behind this failure. In this review, we try to give an overview of the current knowledge about this pathway, to point out the next challenges, and to propose alternative therapeutic routes. Abstract The kynurenine pathway has been highlighted as a gatekeeper of immune-privileged sites through its ability to generate from tryptophan a set of immunosuppressive metabolic intermediates. It additionally constitutes an important source of cellular NAD+ for the organism. Hijacking of its immunosuppressive functions, as recurrently observed in multiple cancers, facilitates immune evasion and promotes tumor development. Based on these observations, researchers have focused on characterizing indoleamine 2,3-dioxygenase (IDO1), the main enzyme catalyzing the first and limiting step of the pathway, and on developing therapies targeting it. Unfortunately, clinical trials studying IDO1 inhibitors have thus far not met expectations, highlighting the need to unravel this complex signaling pathway further. Recent advances demonstrate that these metabolites additionally promote tumor growth, metastatic dissemination and chemoresistance by a combination of paracrine and autocrine effects. Production of NAD+ also contributes to cancer progression by providing cancer cells with enhanced plasticity, invasive properties and chemoresistance. A comprehensive survey of this complexity is challenging but necessary to achieve medical success.
Collapse
|
3
|
Al Abdulsalam EA, Al Harithy RN. Visfatin and global histone H3K9me levels in colon cancer. Ann Med 2021; 53:647-652. [PMID: 34008459 PMCID: PMC8143622 DOI: 10.1080/07853890.2021.1925737] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 04/29/2021] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND Visfatin is considered to be a biomarker in various types of cancers, including colon cancer. Moreover, evidence for epigenetic mechanism must be reported for an association between visfatin level and colon cancer. Therefore, this study was designed to investigate the status of visfatin expression and the global histone three modifications in colon cancerous tissue. METHODS Colon cancerous tissue and paired adjacent non-cancerous tissue from 30 patients were used to determine the global histone three modifications using Western blot. Quantitative real-time polymerase chain reaction (qRT-PCR) was used to assess visfatin's expression level in tissues. RESULTS The visfatin and the global H3K9me expression levels were significantly higher in colon cancerous tissue than in the paired adjacent non-cancerous tissue. CONCLUSION The present study makes a crucial noteworthy contribution to visfatin effect on colon cancer development via H3K9me.
Collapse
Affiliation(s)
- Eman A. Al Abdulsalam
- Department of Biochemistry, College of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Rowyda N. Al Harithy
- Department of Biochemistry, College of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
4
|
Nicotinamide adenine dinucleotide (NAD+): essential redox metabolite, co-substrate and an anti-cancer and anti-ageing therapeutic target. Biochem Soc Trans 2021; 48:733-744. [PMID: 32573651 DOI: 10.1042/bst20190033] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 05/29/2020] [Accepted: 06/01/2020] [Indexed: 01/10/2023]
Abstract
Nicotinamide adenine dinucleotide (NAD+) and its reduced form NADH are essential coupled redox metabolites that primarily promote cellular oxidative (catabolic) metabolic reactions. This enables energy generation through glycolysis and mitochondrial respiration to support cell growth and survival. In addition, many key enzymes that regulate diverse cell functions ranging from gene expression to proteostasis require NAD+ as a co-substrate for their catalytic activity. This includes the NAD+-dependent sirtuin family of protein deacetylases and the PARP family of DNA repair enzymes. Whilst their vital activity consumes NAD+ which is cleaved to nicotinamide, several pathways exist for re-generating NAD+ and sustaining NAD+ homeostasis. However, there is growing evidence of perturbed NAD+ homeostasis and NAD+-regulated processes contributing to multiple disease states. NAD+ levels decline in the human brain and other organs with age and this is associated with neurodegeneration and other age-related diseases. Dietary supplementation with NAD+ precursors is being investigated to counteract this. Paradoxically, many cancers have increased dependency on NAD+. Clinical efforts to exploit this have so far shown limited success. Emerging new opportunities to exploit dysregulation of NAD+ metabolism in cancers are critically discussed. An update is also provided on other key NAD+ research including perturbation of the NAD+ salvage enzyme NAMPT in the context of the tumour microenvironment (TME), methodology to study subcellular NAD+ dynamics in real-time and the regulation of differentiation by competing NAD+ pools.
Collapse
|
5
|
Gu ML, Zhou XX, Ren MT, Shi KD, Yu MS, Jiao WR, Wang YM, Zhong WX, Ji F. Blockage of ETS homologous factor inhibits the proliferation and invasion of gastric cancer cells through the c-Met pathway. World J Gastroenterol 2020; 26:7497-7512. [PMID: 33384550 PMCID: PMC7754554 DOI: 10.3748/wjg.v26.i47.7497] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/13/2020] [Accepted: 11/02/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Gastric cancer (GC) is one of the most common and deadliest types of cancer worldwide due to its delayed diagnosis and high metastatic frequency, but its exact pathogenesis has not been fully elucidated. ETS homologous factor (EHF) is an important member of the ETS family and contributes to the pathogenesis of multiple malignant tumors. To date, whether EHF participates in the development of GC via the c-Met signaling pathway remains unclear.
AIM To investigate the role and mechanism of EHF in the occurrence and development of GC.
METHODS The expression of EHF mRNA in GC tissues and cell lines was measured by quantitative PCR. Western blotting was performed to determine the protein expression of EHF, c-Met, and its downstream signal molecules. The EHF expression in GC tissues was further detected by immunohistochemical staining. To investigate the role of EHF in GC oncogenesis, small interfering RNA (siRNA) against EHF was transfected into GC cells. The cell proliferation of GC cells was determined by Cell Counting Kit-8 and colony formation assays. Flow cytometry was performed following Annexin V/propidium iodide (PI) to identify apoptotic cells and PI staining to analyze the cell cycle. Cell migration and invasion were assessed by transwell assays.
RESULTS The data showed that EHF was upregulated in GC tissues and cell lines in which increased expression of c-Met was also observed. Silencing of EHF by siRNA reduced the proliferation of GC cells. Inhibition of EHF induced significant apoptosis and cell cycle arrest in GC cells. Cell migration and invasion were significantly inhibited. EHF silencing led to c-Met downregulation and further blocked the Ras/c-Raf/extracellular signal-related kinase 1/2 (Erk1/2) pathway. Additionally, phosphatase and tensin homolog was upregulated and glycogen synthase kinase 3 beta was deactivated. Moreover, inactivation of signal transducer and activator of transcription 3 was detected following EHF inhibition, leading to inhibition of the epithelial-to-mesenchymal transition (EMT).
CONCLUSION These results suggest that EHF plays a key role in cell proliferation, invasion, apoptosis, the cell cycle and EMT via the c-Met pathway. Therefore, EHF may serve as an antineoplastic target for the diagnosis and treatment of GC.
Collapse
Affiliation(s)
- Meng-Li Gu
- Department of Gastroenterology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Xin-Xin Zhou
- Department of Gastroenterology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Meng-Ting Ren
- Department of Gastroenterology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Ke-Da Shi
- Department of Gastroenterology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Mo-Sang Yu
- Department of Gastroenterology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Wen-Rui Jiao
- Department of Gastroenterology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Ya-Mei Wang
- Department of Gastroenterology, The Fourth Affiliated Hospital, College of Medicine, Zhejiang University, Yiwu 322000, Zhejiang Province, China
| | - Wei-Xiang Zhong
- Department of Pathology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Feng Ji
- Department of Gastroenterology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| |
Collapse
|
6
|
Tian W, Zhang H, Zhang Y, Wang Y, Zhang Y, Xue F, Song X, Zhang H. High level of visfatin and the activation of Akt and ERK1/2 signaling pathways are associated with endometrium malignant transformation in polycystic ovary syndrome. Gynecol Endocrinol 2020; 36:156-161. [PMID: 31452406 DOI: 10.1080/09513590.2019.1650340] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
This study aimed to assess the clinicopathological significance of serum levels and endometrium tissue expression of visfatin in polycystic ovary syndrome (PCOS) patients. A total of 80 PCOS patients and 80 matching controls were included in this study. We analyzed the relationship between the expression of visfatin in endometrium and clinicopathological characteristics in PCOS patients. The correlation between the expression of visfatin and p-Akt, Akt, p-ERK1/2, and ERK1/2 in PCOS tissues was evaluated as well. Visfatin expression in PCOS endometrial tissues were significantly higher than those in controls (p = .001). The expression of phosphorylation of Akt and ERK1/2 were significantly higher in PCOS endometrium tissues compared to controls (p < .05). Moreover, a high expression of tissue visfatin in PCOS tissues was positively correlated with the expression of p-Akt (p = .015), and p-ERK1/2 (p = .013). Western blotting revealed that protein expression of visfatin in PCOS patients with endometrial hyperplasia and cancer was higher than that in patients with normal endometrium tissues, and the difference was statistically significant (p = .027). The expression of p-Akt (p = .018) and p-ERK1/2 (p = .035) in PCOS patients with endometrial hyperplasia and cancer was significantly higher than that in patients with normal endometrium tissues. Visfatin may be a potential biomarker for endometrial malignant transformation in PCOS patients.
Collapse
Affiliation(s)
- Wenyan Tian
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Huixia Zhang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Yan Zhang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Yingmei Wang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Yanfang Zhang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Fengxia Xue
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Xueru Song
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Huiying Zhang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| |
Collapse
|
7
|
Uchiumi F, Sato A, Asai M, Tanuma SI. An NAD<sup>+</sup> dependent/sensitive transcription system: Toward a novel anti-cancer therapy. AIMS MOLECULAR SCIENCE 2020. [DOI: 10.3934/molsci.2020002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
8
|
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
|
9
|
Lewis JE, Singh N, Holmila RJ, Sumer BD, Williams NS, Furdui CM, Kemp ML, Boothman DA. Targeting NAD + Metabolism to Enhance Radiation Therapy Responses. Semin Radiat Oncol 2019; 29:6-15. [PMID: 30573185 DOI: 10.1016/j.semradonc.2018.10.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Nicotinamide adenine dinucleotide (NAD+) metabolism is integrally connected with the mechanisms of action of radiation therapy and is altered in many radiation-resistant tumors. This makes NAD+ metabolism an ideal target for therapies that increase radiation sensitivity and improve patient outcomes. This review provides an overview of NAD+ metabolism in the context of the cellular response to ionizing radiation, as well as current therapies that target NAD+ metabolism to enhance radiation therapy responses. Additionally, we summarize state-of-the-art methods for measuring, modeling, and manipulating NAD+ metabolism, which are being used to identify novel targets in the NAD+ metabolic network for therapeutic interventions in combination with radiation therapy.
Collapse
Affiliation(s)
- Joshua E Lewis
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA.
| | - Naveen Singh
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
| | - Reetta J Holmila
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - Baran D Sumer
- Departments of Surgery, UT Southwestern Medical Center, Dallas, TX
| | - Noelle S Williams
- Departments of Biochemistry, UT Southwestern Medical Center, Dallas, TX
| | - Cristina M Furdui
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - Melissa L Kemp
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA
| | - David A Boothman
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
10
|
Yaku K, Okabe K, Hikosaka K, Nakagawa T. NAD Metabolism in Cancer Therapeutics. Front Oncol 2018; 8:622. [PMID: 30631755 PMCID: PMC6315198 DOI: 10.3389/fonc.2018.00622] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Accepted: 11/30/2018] [Indexed: 12/15/2022] Open
Abstract
Cancer cells have a unique energy metabolism for sustaining rapid proliferation. The preference for anaerobic glycolysis under normal oxygen conditions is a unique trait of cancer metabolism and is designated as the Warburg effect. Enhanced glycolysis also supports the generation of nucleotides, amino acids, lipids, and folic acid as the building blocks for cancer cell division. Nicotinamide adenine dinucleotide (NAD) is a co-enzyme that mediates redox reactions in a number of metabolic pathways, including glycolysis. Increased NAD levels enhance glycolysis and fuel cancer cells. In fact, nicotinamide phosphoribosyltransferase (Nampt), a rate-limiting enzyme for NAD synthesis in mammalian cells, is frequently amplified in several cancer cells. In addition, Nampt-specific inhibitors significantly deplete NAD levels and subsequently suppress cancer cell proliferation through inhibition of energy production pathways, such as glycolysis, tricarboxylic acid (TCA) cycle, and oxidative phosphorylation. NAD also serves as a substrate for poly(ADP-ribose) polymerase (PARP), sirtuin, and NAD gylycohydrolase (CD38 and CD157); thus, NAD regulates DNA repair, gene expression, and stress response through these enzymes. Thus, NAD metabolism is implicated in cancer pathogenesis beyond energy metabolism and considered a promising therapeutic target for cancer treatment. In this review, we present recent findings with respect to NAD metabolism and cancer pathogenesis. We also discuss the current and future perspectives regarding the therapeutics that target NAD metabolic pathways.
Collapse
Affiliation(s)
- Keisuke Yaku
- Department of Metabolism and Nutrition, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, Toyama, Japan
| | - Keisuke Okabe
- Department of Metabolism and Nutrition, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, Toyama, Japan.,First Department of Internal Medicine, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, Toyama, Japan
| | - Keisuke Hikosaka
- Department of Metabolism and Nutrition, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, Toyama, Japan
| | - Takashi Nakagawa
- Department of Metabolism and Nutrition, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, Toyama, Japan.,Institute of Natural Medicine, University of Toyama, Toyama, Japan
| |
Collapse
|
11
|
Visfatin Promotes Wound Healing through the Activation of ERK1/2 and JNK1/2 Pathway. Int J Mol Sci 2018; 19:ijms19113642. [PMID: 30463229 PMCID: PMC6274809 DOI: 10.3390/ijms19113642] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 11/14/2018] [Accepted: 11/15/2018] [Indexed: 01/20/2023] Open
Abstract
Visfatin, a member of the adipokine family, plays an important role in many metabolic and stress responses. The mechanisms underlying the direct therapeutic effects of visfatin on wound healing have not been reported yet. In this study, we examined the effects of visfatin on wound healing in vitro and in vivo. Visfatin enhanced the proliferation and migration of human dermal fibroblasts (HDFs) and keratinocytes the expression of wound healing-related vascular endothelial growth factor (VEGF) in vitro and in vivo. Treatment of HDFs with visfatin induced activation of both extracellular signal-regulated kinases 1 and 2 (ERK1/2) and c-Jun N-terminal kinases 1 and 2 (JNK1/2) in a time-dependent manner. Inhibition of ERK1/2 and JNK1/2 led to a significant decrease in visfatin-induced proliferation and migration of HDFs. Importantly, blocking VEGF with its neutralizing antibodies suppressed the visfatin-induced proliferation and migration of HDFs and human keratinocytes, indicating that visfatin induces the proliferation and migration of HDFs and human keratinocytes via increased VEGF expression. Moreover, visfatin effectively improved wound repair in vivo, which was comparable to the wound healing activity of epidermal growth factor (EGF). Taken together, we demonstrate that visfatin promotes the proliferation and migration of HDFs and human keratinocytes by inducing VEGF expression and can be used as a potential novel therapeutic agent for wound healing.
Collapse
|
12
|
Li Y, Ke J, Peng C, Wu F, Song Y. microRNA-300/NAMPT regulates inflammatory responses through activation of AMPK/mTOR signaling pathway in neonatal sepsis. Biomed Pharmacother 2018; 108:271-279. [PMID: 30223098 DOI: 10.1016/j.biopha.2018.08.064] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 08/08/2018] [Accepted: 08/15/2018] [Indexed: 01/08/2023] Open
Abstract
AIM Rapid and accurate diagnosis of neonatal sepsis (NS) is highly warranted because of high associated morbidity and mortality. The study aims to evaluate the effects of miR-300 on inflammatory responses in a septic neonate mouse model. METHODS A septic mouse model was established by intraperitoneal (i.p.) cecal slurry (CS) injection in order to validate the effect of miR-300 on the inflammatory response in endothelial cells. Bioinformatics tools and luciferase activity were employed to detect the target of miR-300. Serum inflammatory factors were determined by ELISA assay. RT-qPCR and western blot analysis were used to determine the gene expressions. Flow cytometry was employed to evaluate cell apoptosis. RESULTS Gain- and loss-of-function studies revealed that miR-300 overexpression augmented autophagy, inhibited cell apoptosis, enhanced cell cycle entry in endothelial cells, and decreased inflammatory response through the regulation of pro- and anti-apoptotic factors in endothelial cells. The effect of miR-300on endothelial cells was upregulated after nicotinamide phosphoribosyltransferase (NAMPT) silencing and AMPK/mTOR signaling pathway activation, indicating that miR-300 influences sepsis via suppressing NAMPT and triggering the AMPK/mTOR signaling pathway. CONCLUSIONS Our study provides evidence indicating that overexpressedmiR-300 enhances autophagy by targeting NAMPT through activation of the AMPK/mTOR signaling pathway in septic mouse models, indicating it may serve as a potential therapeutic target for sepsis.
Collapse
Affiliation(s)
- Yexuzi Li
- Department of Medical Intensive Care Unit, The First People's Hospital of Wenling, Wenling 317500, Zhejiang Province, China.
| | - Junzhong Ke
- Department of Medical Intensive Care Unit, The First People's Hospital of Wenling, Wenling 317500, Zhejiang Province, China.
| | - Chen Peng
- Department of Medical Intensive Care Unit, The First People's Hospital of Wenling, Wenling 317500, Zhejiang Province, China.
| | - Fugen Wu
- Department of Neonatal Intensive Care Unit, The First People's Hospital of Wenling, Wenling 317500, Zhejiang Province, China.
| | - Yukang Song
- Department of Medical Intensive Care Unit, The First People's Hospital of Wenling, Wenling 317500, Zhejiang Province, China.
| |
Collapse
|
13
|
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
|
14
|
Sadrerafi K, Mason EO, Lee MW. Clickable prodrugs bearing potent and hydrolytically cleavable nicotinamide phosphoribosyltransferase inhibitors. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:987-995. [PMID: 29731606 PMCID: PMC5923274 DOI: 10.2147/dddt.s152685] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Purpose Our previous study indicated that carborane containing small-molecule 1-(hydroxymethyl)-7-(4′-(trans-3″-(3′″-pyridyl)acrylamido)butyl)-1,7-dicarbadodecaborane (hm-MC4-PPEA), was a potent inhibitor of nicotinamide phosphoribosyltransferase (Nampt). Nampt has been shown to be upregulated in most cancers and is a promising target for the treatment of many different types of cancers, including breast cancers. Patients and methods To increase the selectivity of hm-MC4-PPEA toward cancer cells, three prodrugs were synthesized with different hydrolyzable linkers: ester, carbonate, and carbamate. Using click chemistry a fluorophore was attached to these prodrugs to act as a model for our conjugation strategy and to serve as an aid for prodrug stability studies. The stabilities of these drug conjugates were tested in phosphate-buffered saline (PBS) at normothermia (37°C) using three different pH levels, 5.5, 7.5, and 9.5, as well as in horse serum at physiological pH. The stability of each was monitored using reversed-phase HPLC equipped with both diode array and fluorescence detection. The inhibitory activity of hm-MC4-PPEA was also measured using a commercially available colorimetric assay. The biological activities of the drug conjugates as well as those of the free drug (hm-MC4-PPEA), were evaluated using the MTT assay against the human breast cancer cell lines T47D and MCF7, as well as the noncancerous, transformed, Nampt-dependent human breast epithelium cell line 184A1. Results hm-MC4-PPEA showed to be a potent inhibitor of recombinant Nampt activity, exhibiting an IC50 concentration of 6.8 nM. The prodrugs showed great stability towards hydrolytic degradation under neutral, mildly acidic and mildly basic conditions. The carbamate prodrug also showed to be stable in rat serum. However, the carbonate and the ester prodrug release at various rates in serum presumably owing to the presence of several different classes of esterase. The biological activities of the drug conjugates correlate with the stability of their cleavable linkers observed in serum. Conclusion The targeted and selective delivery of potent Nampt inhibitors to cancer cells is a potentially new route for the treatment of many cancers. These prodrugs linked to small cancer-associated peptides may be optimum for their use as targetable Nampt inhibitors.
Collapse
Affiliation(s)
- Keivan Sadrerafi
- Department of Chemistry, University of Missouri, Columbia, MO, USA
| | - Emilia O Mason
- Department of Chemistry, University of Missouri, Columbia, MO, USA
| | - Mark W Lee
- Department of Chemistry, University of Missouri, Columbia, MO, USA
| |
Collapse
|
15
|
Thongon N, Zucal C, D'Agostino VG, Tebaldi T, Ravera S, Zamporlini F, Piacente F, Moschoi R, Raffaelli N, Quattrone A, Nencioni A, Peyron JF, Provenzani A. Cancer cell metabolic plasticity allows resistance to NAMPT inhibition but invariably induces dependence on LDHA. Cancer Metab 2018. [PMID: 29541451 PMCID: PMC5844108 DOI: 10.1186/s40170-018-0174-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background Inhibitors of nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme in NAD+ biosynthesis from nicotinamide, exhibit anticancer effects in preclinical models. However, continuous exposure to NAMPT inhibitors, such as FK866, can induce acquired resistance. Methods We developed FK866-resistant CCRF-CEM (T cell acute lymphoblastic leukemia) and MDA MB231 (breast cancer) models, and by exploiting an integrated approach based on genetic, biochemical, and genome wide analyses, we annotated the drug resistance mechanisms. Results Acquired resistance to FK866 was independent of NAMPT mutations but rather was based on a shift towards a glycolytic metabolism and on lactate dehydrogenase A (LDHA) activity. In addition, resistant CCRF-CEM cells, which exhibit high quinolinate phosphoribosyltransferase (QPRT) activity, also exploited amino acid catabolism as an alternative source for NAD+ production, becoming addicted to tryptophan and glutamine and sensitive to treatment with the amino acid transport inhibitor JPH203 and with l-asparaginase, which affects glutamine exploitation. Vice versa, in line with their low QPRT expression, FK866-resistant MDA MB231 did not rely on amino acids for their resistance phenotype. Conclusions Our study identifies novel mechanisms of resistance to NAMPT inhibition, which may be useful to design more rational strategies for targeting cancer metabolism.
Collapse
Affiliation(s)
- Natthakan Thongon
- 1Center For Integrative Biology (CIBIO), University of Trento, via Sommarive 9, Trento, Italy
| | - Chiara Zucal
- 1Center For Integrative Biology (CIBIO), University of Trento, via Sommarive 9, Trento, Italy
| | | | - Toma Tebaldi
- 1Center For Integrative Biology (CIBIO), University of Trento, via Sommarive 9, Trento, Italy
| | - Silvia Ravera
- 2Department of Pharmacy, Biochemistry Laboratory, University of Genova, Genova, Italy
| | - Federica Zamporlini
- 3Department of Agricultural, Food and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | | | - Ruxanda Moschoi
- 5Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), INSERM U1065, Nice, France
| | - Nadia Raffaelli
- 3Department of Agricultural, Food and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Alessandro Quattrone
- 1Center For Integrative Biology (CIBIO), University of Trento, via Sommarive 9, Trento, Italy
| | - Alessio Nencioni
- 4Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Jean-Francois Peyron
- 5Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), INSERM U1065, Nice, France
| | - Alessandro Provenzani
- 1Center For Integrative Biology (CIBIO), University of Trento, via Sommarive 9, Trento, Italy
| |
Collapse
|
16
|
Korepanova A, Longenecker KL, Pratt SD, Panchal SC, Clark RF, Lake M, Gopalakrishnan SM, Raich D, Sun C, Petros AM. Fragment-based discovery of a potent NAMPT inhibitor. Bioorg Med Chem Lett 2018; 28:437-440. [DOI: 10.1016/j.bmcl.2017.12.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 12/04/2017] [Accepted: 12/12/2017] [Indexed: 02/07/2023]
|
17
|
Zhao G, Green CF, Hui YH, Prieto L, Shepard R, Dong S, Wang T, Tan B, Gong X, Kays L, Johnson RL, Wu W, Bhattachar S, Del Prado M, Gillig JR, Fernandez MC, Roth KD, Buchanan S, Kuo MS, Geeganage S, Burkholder TP. Discovery of a Highly Selective NAMPT Inhibitor That Demonstrates Robust Efficacy and Improved Retinal Toxicity with Nicotinic Acid Coadministration. Mol Cancer Ther 2017; 16:2677-2688. [PMID: 29054982 DOI: 10.1158/1535-7163.mct-16-0674] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 04/13/2017] [Accepted: 09/21/2017] [Indexed: 11/16/2022]
Abstract
NAMPT, an enzyme essential for NAD+ biosynthesis, has been extensively studied as an anticancer target for developing potential novel therapeutics. Several NAMPT inhibitors have been discovered, some of which have been subjected to clinical investigations. Yet, the on-target hematological and retinal toxicities have hampered their clinical development. In this study, we report the discovery of a unique NAMPT inhibitor, LSN3154567. This molecule is highly selective and has a potent and broad spectrum of anticancer activity. Its inhibitory activity can be rescued with nicotinic acid (NA) against the cell lines proficient, but not those deficient in NAPRT1, essential for converting NA to NAD+ LSN3154567 also exhibits robust efficacy in multiple tumor models deficient in NAPRT1. Importantly, this molecule when coadministered with NA does not cause observable retinal and hematological toxicities in the rodents, yet still retains robust efficacy. Thus, LSN3154567 has the potential to be further developed clinically into a novel cancer therapeutic. Mol Cancer Ther; 16(12); 2677-88. ©2017 AACR.
Collapse
Affiliation(s)
- Genshi Zhao
- Cancer Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana.
| | - Colin F Green
- Toxicology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | - Yu-Hua Hui
- Drug Disposition, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | - Lourdes Prieto
- Discovery Chemistry Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | - Robert Shepard
- Cancer Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | - Sucai Dong
- Cancer Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | - Tao Wang
- Cancer Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | - Bo Tan
- Toxicology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | - Xueqian Gong
- Cancer Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | - Lisa Kays
- Cancer Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | - Robert L Johnson
- Toxicology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | - Wenjuan Wu
- Cancer Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | - Shobha Bhattachar
- Product Design and Developability, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | - Miriam Del Prado
- Discovery Chemistry Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | - James R Gillig
- Discovery Chemistry Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | - Maria-Carmen Fernandez
- Discovery Chemistry Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | - Ken D Roth
- Discovery Chemistry Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | - Sean Buchanan
- Cancer Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | - Ming-Shang Kuo
- Discovery Chemistry Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | - Sandaruwan Geeganage
- Cancer Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | - Timothy P Burkholder
- Discovery Chemistry Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| |
Collapse
|
18
|
Visfatin mediates doxorubicin resistance in human colorectal cancer cells via up regulation of multidrug resistance 1 (MDR1). Cancer Chemother Pharmacol 2017; 80:395-403. [PMID: 28667355 DOI: 10.1007/s00280-017-3365-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 06/15/2017] [Indexed: 10/19/2022]
Abstract
Colorectal cancer (CRC) is one of the prevalent and deadly cancers worldwide. Chemotherapy resistance is one of the most challenging problems for CRC and other cancer treatments. Recent studies indicated that increasing levels of visfatin are correlated with worse clinical prognosis of CRC patients, while the effects and mechanisms of visfatin on progression of CRC remain unclear. Our present study established doxorubicin (Dox)-resistant CRC HCT-116 and SW480 cells (named HCT-116 Dox/R and SW480 Dox/R). The expression of visfatin, while not IL-6, IL-8, or TGF-β, in CRC Dox-resistant cells was significantly greater than that in their parental cells, while knockdown of visfatin by its specific siRNAs can elevate Dox sensitivity of CRC-resistant cells. In addition, si-visfatin can significantly down regulate the expression of multidrug resistance 1 (MDR1), while not multidrug resistance-associated protein 1 or lung resistance-related protein, in both HCT-116 Dox/R and SW480 Dox/R cells. Visfatin can regulate the transcription of MDR1 via modulating its promoter activities. Si-visfatin can also decrease the activation and nuclear localization of p65, one of the most important transcription factors for the expression of MDR1. Chromatin immunoprecipitation (ChIP) indicated that si-visfatin can suppress the binding between p65 and MDR1 promoter. Collectively, our present study revealed that visfatin mediates the Dox resistance of CRC cells via up regulation of MDR1. It indicated that targeted inhibition of visfatin might be helpful for overcoming Dox resistance of CRC therapy.
Collapse
|
19
|
Dalamaga M, Christodoulatos GS. Visfatin, Obesity, and Cancer. ADIPOCYTOKINES, ENERGY BALANCE, AND CANCER 2017. [DOI: 10.1007/978-3-319-41677-9_6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
20
|
Inhibitor of Nicotinamide Phosphoribosyltransferase Sensitizes Glioblastoma Cells to Temozolomide via Activating ROS/JNK Signaling Pathway. BIOMED RESEARCH INTERNATIONAL 2016; 2016:1450843. [PMID: 28097126 PMCID: PMC5206411 DOI: 10.1155/2016/1450843] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 11/10/2016] [Indexed: 12/16/2022]
Abstract
Overcoming temozolomide (TMZ) resistance is a great challenge in glioblastoma (GBM) treatment. Nicotinamide phosphoribosyltransferase (NAMPT) is a rate-limiting enzyme in the biosynthesis of nicotinamide adenine dinucleotide and has a crucial role in cancer cell metabolism. In this study, we investigated whether FK866 and CHS828, two specific NAMPT inhibitors, could sensitize GBM cells to TMZ. Low doses of FK866 and CHS828 (5 nM and 10 nM, resp.) alone did not significantly decrease cell viability in U251-MG and T98 GBM cells. However, they significantly increased the antitumor action of TMZ in these cells. In U251-MG cells, administration of NAMPT inhibitors increased the TMZ (100 μM)-induced apoptosis and LDH release from GBM cells. NAMPT inhibitors remarkably enhanced the activities of caspase-1, caspase-3, and caspase-9. Moreover, NAMPT inhibitors increased reactive oxygen species (ROS) production and superoxide anion level but reduced the SOD activity and total antioxidative capacity in GBM cells. Treatment of NAMPT inhibitors increased phosphorylation of c-Jun and JNK. Administration of JNK inhibitor SP600125 or ROS scavenger tocopherol with TMZ and NAMPT inhibitors substantially attenuated the sensitization of NAMPT inhibitor on TMZ antitumor action. Our data indicate a potential value of NAMPT inhibitors in combined use with TMZ for GBM treatment.
Collapse
|
21
|
Improved synthesis of MC4-PPEA and the biological evaluation of its hydroxymethyl derivative. Bioorg Med Chem Lett 2016; 26:618-621. [DOI: 10.1016/j.bmcl.2015.11.068] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 11/13/2015] [Accepted: 11/19/2015] [Indexed: 11/23/2022]
|
22
|
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: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 09/23/2015] [Indexed: 11/05/2022] Open
|
23
|
Copy number variants associated with 18p11.32, DCC and the promoter 1B region of APC in colorectal polyposis patients. Meta Gene 2015; 7:95-104. [PMID: 26909336 PMCID: PMC4733217 DOI: 10.1016/j.mgene.2015.12.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 12/16/2015] [Accepted: 12/21/2015] [Indexed: 01/05/2023] Open
Abstract
Familial Adenomatous Polyposis (FAP) is the second most common inherited predisposition to colorectal cancer (CRC) associated with the development of hundreds to thousands of adenomas in the colon and rectum. Mutations in APC are found in ~ 80% polyposis patients with FAP. In the remaining 20% no genetic diagnosis can be provided suggesting other genes or mechanisms that render APC inactive may be responsible. Copy number variants (CNVs) remain to be investigated in FAP and may account for disease in a proportion of polyposis patients. A cohort of 56 polyposis patients and 40 controls were screened for CNVs using the 2.7M microarray (Affymetrix) with data analysed using ChAS (Affymetrix). A total of 142 CNVs were identified unique to the polyposis cohort suggesting their involvement in CRC risk. We specifically identified CNVs in four unrelated polyposis patients among CRC susceptibility genes APC, DCC, MLH1 and CTNNB1 which are likely to have contributed to disease development in these patients. A recurrent deletion was observed at position 18p11.32 in 9% of the patients screened that was of particular interest. Further investigation is necessary to fully understand the role of these variants in CRC risk given the high prevalence among the patients screened.
Collapse
Key Words
- ALL, acute lymphoblastic leukaemia
- BH, Bengamini and Hochberg
- CHAS, Chromosome Analysis Suite
- CN, copy number
- CNV
- CNV, copy number variation
- COSMIC, Catalogue of Somatic Mutations in Cancer
- CRC, colorectal cancer
- Cancer
- DGV, Database of genomic variants
- DNA, deoxyribose nucleic acid
- FAP, familial adenomatous polyposis
- HMDD, human microRNA disease database
- KEGG, Kyoto Encyclopaedia of Genes and Genomes
- Kb, kilobase
- LOH, loss of heterozygosity
- MLPA, multiplex ligation-dependant probe amplification
- MMR, mismatch repair
- NTC, no template control
- QC, quality control
- RNA, ribose nucleic acid
- SNP, single nucleotide polymorphism
- TAM, Tool for the annotation of microRNAs
- TCGA, The Cancer Genome Atlas
- UCSC, University of California, Santa Cruz
- diagnostic testing
- lncRNA, link RNA
- long non-coding RNAs
- mapd, median absolute pairwise difference
- miR, microRNA
- ng, nanogram
- polyposis
Collapse
|
24
|
Ezeoke CC, Morley JE. Pathophysiology of anorexia in the cancer cachexia syndrome. J Cachexia Sarcopenia Muscle 2015; 6:287-302. [PMID: 26675762 PMCID: PMC4670736 DOI: 10.1002/jcsm.12059] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 06/11/2015] [Accepted: 06/22/2015] [Indexed: 12/20/2022] Open
Abstract
Anorexia is commonly present in persons with cancer and a major component of cancer cachexia. There are multiple causes of anorexia in cancer. Peripherally, these can be due to (i) substances released from or by the tumour, e.g. pro-inflammatory cytokines, lactate, and parathormone-related peptide; (ii) tumours causing dysphagia or altering gut function; (iii) tumours altering nutrients, e.g. zinc deficiency; (iv) tumours causing hypoxia; (v) increased peripheral tryptophan leading to increased central serotonin; or (vi) alterations of release of peripheral hormones that alter feeding, e.g. peptide tyrosine tyrosine and ghrelin. Central effects include depression and pain, decreasing the desire to eat. Within the central nervous system, tumours create multiple alterations in neurotransmitters, neuropeptides, and prostaglandins that modulate feeding. Many of these neurotransmitters appear to produce their anorectic effects through the adenosine monophosphate kinase/methylmalonyl coenzyme A/fatty acid system in the hypothalamus. Dynamin is a guanosine triphosphatase that is responsible for internalization of melanocortin 4 receptors and prostaglandin receptors. Dynamin is up-regulated in a mouse model of cancer anorexia. A number of drugs, e.g. megestrol acetate, cannabinoids, and ghrelin agonists, have been shown to have some ability to be orexigenic in cancer patients.
Collapse
Affiliation(s)
- Chukwuemeka Charles Ezeoke
- United States Navy Medical Corps and PGY-2, Internal Medicine Residency, Saint Louis University HospitalSt. Louis, MO, USA
| | - John E Morley
- Division of Geriatrics, Saint Louis University School of Medicine1402 S. Grand Blvd., M238, St. Louis, MO, 63104, USA
- Division of Endocrinology, Saint Louis University School of MedicineSt. Louis, MO, USA
| |
Collapse
|
25
|
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
|
26
|
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
|
27
|
Cheng G, Liu C, Sun X, Zhang L, Liu L, Ouyang J, Li B. Visfatin promotes osteosarcoma cell migration and invasion via induction of epithelial-mesenchymal transition. Oncol Rep 2015; 34:987-94. [PMID: 26062797 DOI: 10.3892/or.2015.4053] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Accepted: 04/02/2015] [Indexed: 11/06/2022] Open
Abstract
Visfatin is considered to be a biomarker in various types of cancers. However, no evidence has been reported for the direct effect of visfatin on osteosarcoma cell metastasis. The aims of the present study were to investigate the influence of visfatin on the migration and invasion of osteosarcoma cells and clarify the underlying mechanism. The expression levels of epithelial-mesenchymal transition (EMT) markers, as well as the transcriptional factor Snail-1, were first detected at both the protein and mRNA levels in U2OS osteosarcoma cells after stimulation of visfatin. Then the expression of NF-κB (p65) was detected by western blot analysis, and siRNA of Snail-1 and inhibitor of NF-κB were used to investigate the effect of visfatin. Finally, migration and invasion of the cells were detected respectively by scratch wound healing and transwell assays. Visfatin downregulated E-cadherin and upregulated N-cadherin in concentration- and time-dependent manners at the protein and mRNA levels. The expression of Snail-1 was also upregulated. Moreover, visfatin also promoted the nuclear translocation of the NF-κB pathway. Administration of siRNA of Snail-1 and the inhibitor BAY11-7082 validated the roles of Snail-1 and NF-κB in the visfatin-induced regulation of EMT markers. Migration and invasion of U2OS osteosarcoma cells were promoted following the application of visfatin. These results demonstrated that visfatin enhances the migration and invasion of osteosarcoma cells via the NF-κB/Snail-1/EMT pathway.
Collapse
Affiliation(s)
- Gong Cheng
- Department of Anatomy, Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Changying Liu
- Department of Orthopedics, People's Hospital of Linyi City, Linyi, Shandong, P.R. China
| | - Xiujiang Sun
- Department of Sports Medicine, Yantaishan Hospital, Yantai, Shandong, P.R. China
| | - Lei Zhang
- Department of Gastroenterology, Central Hospital of Zibo, Zibo, Shandong, P.R. China
| | - Lifang Liu
- Department of Cardiology, Central Hospital of Zibo, Zibo, Shandong, P.R. China
| | - Jun Ouyang
- Department of Anatomy, Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Bo Li
- Department of Cardiology, Central Hospital of Zibo, Zibo, Shandong, P.R. China
| |
Collapse
|
28
|
Nampt/PBEF/visfatin upregulation in colorectal tumors, mirrored in normal tissue and whole blood of colorectal cancer patients, is associated with metastasis, hypoxia, IL1β, and anemia. BIOMED RESEARCH INTERNATIONAL 2015; 2015:523930. [PMID: 26075243 PMCID: PMC4444566 DOI: 10.1155/2015/523930] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 04/20/2015] [Accepted: 04/23/2015] [Indexed: 12/30/2022]
Abstract
Targeting Nampt/PBEF/visfatin is considered a promising anticancer strategy, yet little is known about its association with colorectal cancer (CRC). We quantified Nampt/PBEF/visfatin expression in bowel and blood (mRNA and protein), referring it to CRC advancement and inflammatory, angiogenic, hypoxia, and proliferation indices. Tumor Nampt/PBEF/visfatin upregulation was associated with metastasis, anemia, tumor location, HIF1α, and inflammatory and angiogenic indices, of which HIF1α, IL1β, and anemia explained 70% in Nampt/PBEF/visfatin variability. Nampt/PBEF/visfatin expression in nontumor tissue, both mRNA and protein, increased in patients with metastatic disease and mild anemia, and, on transcriptional level, correlated with HIF1α, IL1β, IL8, CCL2, and CCL4 expression. Whole blood Nampt/PBEF/visfatin tended to be elevated in patients with metastatic cancer or anemia and correlated with inflammatory indices, of which IL1β, IL8, and hematocrit explained 60% of its variability. Circulating visfatin was associated with lymph node metastasis and inflammatory and angiogenic indices. In vitro experiments on SW620 cells demonstrated Nampt/PBEF/visfatin downregulation in response to serum withdrawal but its upregulation in response to serum induction and hypoxia. Stimulation with recombinant visfatin did not provide growth advantage. Summarizing, our results link Nampt/PBEF/visfatin with tumor metastatic potential and point at inflammation and hypoxia as key inducers of its upregulation in CRC.
Collapse
|
29
|
Tan B, Dong S, Shepard RL, Kays L, Roth KD, Geeganage S, Kuo MS, Zhao G. Inhibition of Nicotinamide Phosphoribosyltransferase (NAMPT), an Enzyme Essential for NAD+ Biosynthesis, Leads to Altered Carbohydrate Metabolism in Cancer Cells. J Biol Chem 2015; 290:15812-15824. [PMID: 25944913 DOI: 10.1074/jbc.m114.632141] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Indexed: 12/18/2022] Open
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) has been extensively studied due to its essential role in NAD(+) biosynthesis in cancer cells and the prospect of developing novel therapeutics. To understand how NAMPT regulates cellular metabolism, we have shown that the treatment with FK866, a specific NAMPT inhibitor, leads to attenuation of glycolysis by blocking the glyceraldehyde 3-phosphate dehydrogenase step (Tan, B., Young, D. A., Lu, Z. H., Wang, T., Meier, T. I., Shepard, R. L., Roth, K., Zhai, Y., Huss, K., Kuo, M. S., Gillig, J., Parthasarathy, S., Burkholder, T. P., Smith, M. C., Geeganage, S., and Zhao, G. (2013) Pharmacological inhibition of nicotinamide phosphoribosyltransferase (NAMPT), an enzyme essential for NAD(+) biosynthesis, in human cancer cells: metabolic basis and potential clinical implications. J. Biol. Chem. 288, 3500-3511). Due to technical limitations, we failed to separate isotopomers of phosphorylated sugars. In this study, we developed an enabling LC-MS methodology. Using this, we confirmed the previous findings and also showed that NAMPT inhibition led to accumulation of fructose 1-phosphate and sedoheptulose 1-phosphate but not glucose 6-phosphate, fructose 6-phosphate, and sedoheptulose 7-phosphate as previously thought. To investigate the metabolic basis of the metabolite formation, we carried out biochemical and cellular studies and established the following. First, glucose-labeling studies indicated that fructose 1-phosphate was derived from dihydroxyacetone phosphate and glyceraldehyde, and sedoheptulose 1-phosphate was derived from dihydroxyacetone phosphate and erythrose via an aldolase reaction. Second, biochemical studies showed that aldolase indeed catalyzed these reactions. Third, glyceraldehyde- and erythrose-labeling studies showed increased incorporation of corresponding labels into fructose 1-phosphate and sedoheptulose 1-phosphate in FK866-treated cells. Fourth, NAMPT inhibition led to increased glyceraldehyde and erythrose levels in the cell. Finally, glucose-labeling studies showed accumulated fructose 1,6-bisphosphate in FK866-treated cells mainly derived from dihydroxyacetone phosphate and glyceraldehyde 3-phosphate. Taken together, this study shows that NAMPT inhibition leads to attenuation of glycolysis, resulting in further perturbation of carbohydrate metabolism in cancer cells. The potential clinical implications of these findings are also discussed.
Collapse
Affiliation(s)
- Bo Tan
- Discovery Chemistry, Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana 46285
| | - Sucai Dong
- Cancer Research, Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana 46285
| | - Robert L Shepard
- Cancer Research, Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana 46285
| | - Lisa Kays
- Cancer Research, Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana 46285
| | - Kenneth D Roth
- Discovery Chemistry, Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana 46285
| | - Sandaruwan Geeganage
- Cancer Research, Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana 46285
| | - Ming-Shang Kuo
- Discovery Chemistry, Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana 46285.
| | - Genshi Zhao
- Cancer Research, Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana 46285.
| |
Collapse
|
30
|
Zhang K, Zhou B, Zhang P, Zhang Z, Chen P, Pu Y, Song Y, Zhang L. Prognostic value of serum nicotinamide phosphoribosyltransferase in patients with bladder cancer. Croat Med J 2015; 55:507-13. [PMID: 25358883 PMCID: PMC4228298 DOI: 10.3325/cmj.2014.55.507] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Aim To analyze the serum nicotinamide phosphoribosyltransferase (Nampt) level and its prognostic value in bladder cancer (BC). Methods The study included 131 patients with transitional cell BC and 109 healthy controls from the West China Hospital of Sichuan University in the period between 2007 and 2013. Nampt concentration in serum was measured by commercial ELISA kits for human Nampt. Results The serum Nampt protein level in patients with BC (mean ± standard deviation, 16.02 ± 7.95 ng/mL) was significantly higher than in the control group (6.46 ± 2.08 ng/mL) (P < 0.001). Serum Nampt level was an independent prognostic marker of non-muscle-invasive BC, with a higher serum Nampt level (>14.74 ng/mL) indicating shorter recurrence-free survival rate (hazard ratio = 2.85, 95% confidence interval, 1.01-8.06; P = 0.048). Conclusion Our results suggest that serum Nampt level may serve as a biomarker of BC and an independent prognostic marker of non-muscle-invasive BC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Lin Zhang
- Lin Zhang, Department of Forensic Biology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu 610041, PR China,
| |
Collapse
|
31
|
Chakrabarti G, Gerber DE, Boothman DA. Expanding antitumor therapeutic windows by targeting cancer-specific nicotinamide adenine dinucleotide phosphate-biogenesis pathways. Clin Pharmacol 2015; 7:57-68. [PMID: 25870517 PMCID: PMC4381889 DOI: 10.2147/cpaa.s79760] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Nicotinamide adenine dinucleotide phosphate (NADPH) biogenesis is an essential mechanism by which both normal and cancer cells maintain redox balance. While antitumor approaches to treat cancers through elevated reactive oxygen species (ROS) are not new ideas, depleting specific NADPH-biogenesis pathways that control recovery and repair pathways are novel, viable approaches to enhance cancer therapy. However, to elicit efficacious therapies exploiting NADPH-biogenic pathways, it is crucial to understand and specifically define the roles of NADPH-biogenesis pathways used by cancer cells for survival or recovery from cell stress. It is equally important to select NADPH-biogenic pathways that are expendable or not utilized in normal tissue to avoid unwanted toxicity. Here, we address recent literature that demonstrates specific tumor-selective NADPH-biogenesis pathways that can be exploited using agents that target specific cancer cell pathways normally not utilized in normal cells. Defining NADPH-biogenesis profiles of specific cancer-types should enable novel strategies to exploit these therapeutic windows for increased efficacy against recalcitrant neoplastic disease, such as pancreatic cancers. Accomplishing the goal of using ROS as a weapon against cancer cells will also require agents, such as NQO1 bioactivatable drugs, that selectively induce elevated ROS levels in cancer cells, while normal cells are protected.
Collapse
Affiliation(s)
- Gaurab Chakrabarti
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, USA ; Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, TX, USA ; Harold C Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA
| | - David E Gerber
- Division of Hematology and Oncology, UT Southwestern Medical Center, Dallas, TX, USA ; Harold C Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA
| | - David A Boothman
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, USA ; Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, TX, USA ; Harold C Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
32
|
Metabolomics analysis of metabolic effects of nicotinamide phosphoribosyltransferase (NAMPT) inhibition on human cancer cells. PLoS One 2014; 9:e114019. [PMID: 25486521 PMCID: PMC4259317 DOI: 10.1371/journal.pone.0114019] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 11/04/2014] [Indexed: 01/21/2023] Open
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) plays an important role in cellular bioenergetics. It is responsible for converting nicotinamide to nicotinamide adenine dinucleotide, an essential molecule in cellular metabolism. NAMPT has been extensively studied over the past decade due to its role as a key regulator of nicotinamide adenine dinucleotide–consuming enzymes. NAMPT is also known as a potential target for therapeutic intervention due to its involvement in disease. In the current study, we used a global mass spectrometry–based metabolomic approach to investigate the effects of FK866, a small molecule inhibitor of NAMPT currently in clinical trials, on metabolic perturbations in human cancer cells. We treated A2780 (ovarian cancer) and HCT-116 (colorectal cancer) cell lines with FK866 in the presence and absence of nicotinic acid. Significant changes were observed in the amino acids metabolism and the purine and pyrimidine metabolism. We also observed metabolic alterations in glycolysis, the citric acid cycle (TCA), and the pentose phosphate pathway. To expand the range of the detected polar metabolites and improve data confidence, we applied a global metabolomics profiling platform by using both non-targeted and targeted hydrophilic (HILIC)-LC-MS and GC-MS analysis. We used Ingenuity Knowledge Base to facilitate the projection of metabolomics data onto metabolic pathways. Several metabolic pathways showed differential responses to FK866 based on several matches to the list of annotated metabolites. This study suggests that global metabolomics can be a useful tool in pharmacological studies of the mechanism of action of drugs at a cellular level.
Collapse
|
33
|
Visfatin/pre-B cell colony-enhancing factor immunohistochemical overexpression in oral cancers. J Appl Biomed 2014. [DOI: 10.1016/j.jab.2014.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
|
34
|
The function of nicotinamide phosphoribosyltransferase in the heart. DNA Repair (Amst) 2014; 23:64-8. [PMID: 25277684 DOI: 10.1016/j.dnarep.2014.08.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 05/30/2014] [Accepted: 08/28/2014] [Indexed: 02/07/2023]
Abstract
In addition to its roles as a coenzyme and an electron transfer molecule, nicotinamide adenine dinucleotide (NAD+) has emerged as a substrate of sirtuins, a family of enzymes that control aging and metabolism. Nicotinamide phosphoribosyltransferase (Nampt), a rate-limiting enzyme in the NAD+ salvage pathway, plays an important role in controlling the level of NAD+ and the activity of Sirt1 in the heart and the cardiomyocytes therein. Nampt protects the heart from ischemia and reperfusion injury by stimulating Sirt1. In this review, we summarize what is currently known regarding the function of Nampt in the heart.
Collapse
|
35
|
Bułdak RJ, Skonieczna M, Bułdak Ł, Matysiak N, Mielańczyk Ł, Wyrobiec G, Kukla M, Michalski M, Żwirska-Korczala K. Changes in subcellular localization of visfatin in human colorectal HCT-116 carcinoma cell line after cytochalasin B treatment. Eur J Histochem 2014; 58:2408. [PMID: 25308845 PMCID: PMC4194393 DOI: 10.4081/ejh.2014.2408] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 06/23/2014] [Accepted: 06/24/2014] [Indexed: 12/27/2022] Open
Abstract
The aim of the study was to assess the expression and subcellular localization of visfatin in HCT-116 colorectal carcinoma cells after cytokinesis failure using Cytochalasin B (CytB) and the mechanism of apoptosis of cells after CytB. We observed translocation of visfatin’s antigen in cytB treated colorectal carcinoma HCT-116 cells from cytosol to nucleus. Statistical and morphometric analysis revealed significantly higher area-related numerical density visfatin-bound nano-golds in the nuclei of cytB-treated HCT-116 cells compared to cytosol. Reverse relation to visfatin subcellular localization was observed in un-treated HCT-116 cells. The total amount of visfatin protein and visfatin mRNA level in HCT-116 cells was also decreased after CytB treatment. Additionally, CytB significantly decreased cell survival, increased levels of G2/M fractions, induced bi-nuclei formation as well as increased reactive oxygen species (ROS) level in HCT-116 cells. CytB treatment showed cytotoxic effect that stem from oxidative stress and is connected with the changes in the cytoplasmic/nuclear amount of visfatin in HCT-116 cells.
Collapse
|
36
|
Neumayer G, Belzil C, Gruss OJ, Nguyen MD. TPX2: of spindle assembly, DNA damage response, and cancer. Cell Mol Life Sci 2014; 71:3027-47. [PMID: 24556998 PMCID: PMC11114040 DOI: 10.1007/s00018-014-1582-7] [Citation(s) in RCA: 147] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 01/28/2014] [Accepted: 01/30/2014] [Indexed: 10/25/2022]
Abstract
For more than 15 years, TPX2 has been studied as a factor critical for mitosis and spindle assembly. These functions of TPX2 are attributed to its Ran-regulated microtubule-associated protein properties and to its control of the Aurora A kinase. Overexpressed in cancers, TPX2 is being established as marker for the diagnosis and prognosis of malignancies. During interphase, TPX2 resides preferentially in the nucleus where its function had remained elusive until recently. The latest finding that TPX2 plays a role in amplification of the DNA damage response, combined with the characterization of TPX2 knockout mice, open new perspectives to understand the biology of this protein. This review provides an historic overview of the discovery of TPX2 and summarizes its cytoskeletal and signaling roles with relevance to cancer therapies. Finally, the review aims to reconcile discrepancies between the experimental and pathological effects of TPX2 overexpression and advances new roles for compartmentalized TPX2.
Collapse
Affiliation(s)
- Gernot Neumayer
- Department of Clinical Neurosciences, Department of Cell Biology and Anatomy, Department of Biochemistry and Molecular Biology, Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, T2N 4N1, Canada,
| | | | | | | |
Collapse
|
37
|
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
|
38
|
Oh A, Ho YC, Zak M, Liu Y, Chen X, Yuen PW, Zheng X, Liu Y, Dragovich PS, Wang W. Structural and biochemical analyses of the catalysis and potency impact of inhibitor phosphoribosylation by human nicotinamide phosphoribosyltransferase. Chembiochem 2014; 15:1121-30. [PMID: 24797455 DOI: 10.1002/cbic.201402023] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Indexed: 11/11/2022]
Abstract
Prolonged inhibition of nicotinamide phosphoribosyltransferase (NAMPT) is a strategy for targeting cancer metabolism. Many NAMPT inhibitors undergo NAMPT-catalyzed phosphoribosylation (pRib), a property often correlated with their cellular potency. To understand this phenomenon and facilitate drug design, we analyzed a potent cellularly active NAMPT inhibitor (GNE-617). A crystal structure of pRib-GNE-617 in complex with NAMPT protein revealed a relaxed binding mode. Consistently, the adduct formation resulted in tight binding and strong product inhibition. In contrast, a biochemically equipotent isomer of GNE-617 (GNE-643) also formed pRib adducts but displayed significantly weaker cytotoxicity. Structural analysis revealed an altered ligand conformation of GNE-643, thus suggesting weak association of the adducts with NAMPT. Our data support a model for cellularly active NAMPT inhibitors that undergo NAMPT-catalyzed phosphoribosylation to produce pRib adducts that retain efficient binding to the enzyme.
Collapse
Affiliation(s)
- Angela Oh
- Genentech, Inc., Department of Structural Biology, 1 DNA Way, South San Francisco, California 94080 (USA)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Ivyna Bong PN, Ng CC, Lam KY, Megat Baharuddin PJN, Chang KM, Zakaria Z. Identification of novel pathogenic copy number aberrations in multiple myeloma: the Malaysian context. Mol Cytogenet 2014; 7:24. [PMID: 24690091 PMCID: PMC4021726 DOI: 10.1186/1755-8166-7-24] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 03/24/2014] [Indexed: 02/07/2023] Open
Abstract
Background Multiple myeloma is an incurable disease. Little is known about the genetic and molecular mechanisms governing the pathogenesis of multiple myeloma. The risk of multiple myeloma predispositions varies among different ethnicities. More than 50% of myeloma cases showed normal karyotypes with conventional cytogenetic analysis due to the low mitotic activity and content of plasma cells in the bone marrow. In the present study, high resolution array comparative genomic hybridization technique was used to identify copy number aberrations in 63 multiple myeloma patients of Malaysia. Results Copy number aberrations were identified in 100% of patients analyzed (n = 63). Common chromosomal gains were detected at regions 1q, 2q, 3p, 3q, 4q, 5q, 6q, 8q, 9q, 10q, 11q, 13q, 14q, 15q, 21q and Xq while common chromosomal losses were identified at regions 3q and 14q. There were a total of 25 and 5 genes localized within the regions of copy number gains and losses, respectively (>30% penetrance). The LYST, CLK1, ACSL1 and NFKBIA are genes localized within the copy number aberration regions and they represent novel information that has never been previously described in multiple myeloma patients. Conclusions In general, due to the differences in genetic background, dietary and lifestyle practices of Malaysian compared to the Caucasian population, these chromosomal alterations might be unique for Asian MM patients. Genes identified in this study could be potential molecular therapeutic targets for the treatment and management of patients with multiple myeloma.
Collapse
Affiliation(s)
- Pau Ni Ivyna Bong
- Hematology Unit, Cancer Research Centre, Institute for Medical Research, Kuala Lumpur, Malaysia.
| | | | | | | | | | | |
Collapse
|
40
|
Schuster S, Penke M, Gorski T, Petzold-Quinque S, Damm G, Gebhardt R, Kiess W, Garten A. Resveratrol differentially regulates NAMPT and SIRT1 in Hepatocarcinoma cells and primary human hepatocytes. PLoS One 2014; 9:e91045. [PMID: 24603648 PMCID: PMC3946349 DOI: 10.1371/journal.pone.0091045] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 02/09/2014] [Indexed: 02/08/2023] Open
Abstract
Resveratrol is reported to possess chemotherapeutic properties in several cancers. In this study, we wanted to investigate the molecular mechanisms of resveratrol-induced cell cycle arrest and apoptosis as well as the impact of resveratrol on NAMPT and SIRT1 protein function and asked whether there are differences in hepatocarcinoma cells (HepG2, Hep3B cells) and non-cancerous primary human hepatocytes. We found a lower basal NAMPT mRNA and protein expression in hepatocarcinoma cells compared to primary hepatocytes. In contrast, SIRT1 was significantly higher expressed in hepatocarcinoma cells than in primary hepatocytes. Resveratrol induced cell cycle arrest in the S- and G2/M- phase and apoptosis was mediated by activation of p53 and caspase-3 in HepG2 cells. In contrast to primary hepatocytes, resveratrol treated HepG2 cells showed a reduction of NAMPT enzymatic activity and increased p53 acetylation (K382). Resveratrol induced NAMPT release from HepG2 cells which was associated with increased NAMPT mRNA expression. This effect was absent in primary hepatocytes where resveratrol was shown to function as NAMPT and SIRT1 activator. SIRT1 inhibition by EX527 resembled resveratrol effects on HepG2 cells. Furthermore, a SIRT1 overexpression significantly decreased both p53 hyperacetylation and resveratrol-induced NAMPT release as well as S-phase arrest in HepG2 cells. We could show that NAMPT and SIRT1 are differentially regulated by resveratrol in hepatocarcinoma cells and primary hepatocytes and that resveratrol did not act as a SIRT1 activator in hepatocarcinoma cells.
Collapse
Affiliation(s)
- Susanne Schuster
- Center for Pediatric Research Leipzig, University Hospital for Children and Adolescents, Faculty of Medicine, University of Leipzig, Leipzig, Germany
- * E-mail:
| | - Melanie Penke
- Center for Pediatric Research Leipzig, University Hospital for Children and Adolescents, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Theresa Gorski
- Center for Pediatric Research Leipzig, University Hospital for Children and Adolescents, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Stefanie Petzold-Quinque
- Center for Pediatric Research Leipzig, University Hospital for Children and Adolescents, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Georg Damm
- Department of General-, Visceral- and Transplantation Surgery, Charité University Medicine Berlin, Berlin, Germany
| | - Rolf Gebhardt
- Institute of Biochemistry, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Wieland Kiess
- Center for Pediatric Research Leipzig, University Hospital for Children and Adolescents, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Antje Garten
- Center for Pediatric Research Leipzig, University Hospital for Children and Adolescents, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| |
Collapse
|
41
|
Ginet V, Puyal J, Rummel C, Aubry D, Breton C, Cloux AJ, Majjigapu SR, Sordat B, Vogel P, Bruzzone S, Nencioni A, Duchosal MA, Nahimana A. A critical role of autophagy in antileukemia/lymphoma effects of APO866, an inhibitor of NAD biosynthesis. Autophagy 2014; 10:603-17. [PMID: 24487122 DOI: 10.4161/auto.27722] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
APO866, an inhibitor of NAD biosynthesis, exhibits potent antitumor properties in various malignancies. Recently, it has been shown that APO866 induces apoptosis and autophagy in human hematological cancer cells, but the role of autophagy in APO866-induced cell death remains unclear. Here, we report studies on the molecular mechanisms underlying APO866-induced cell death with emphasis on autophagy. Treatment of leukemia and lymphoma cells with APO866 induced both autophagy, as evidenced by an increase in autophagosome formation and in SQSTM1/p62 degradation, but also increased caspase activation as revealed by CASP3/caspase 3 cleavage. As an underlying mechanism, APO866-mediated autophagy was found to deplete CAT/catalase, a reactive oxygen species (ROS) scavenger, thus promoting ROS production and cell death. Inhibition of autophagy by ATG5 or ATG7 silencing prevented CAT degradation, ROS production, caspase activation, and APO866-induced cell death. Finally, supplementation with exogenous CAT also abolished APO866 cytotoxic activity. Altogether, our results indicated that autophagy is essential for APO866 cytotoxic activity on cells from hematological malignancies and also indicate an autophagy-dependent CAT degradation, a novel mechanism for APO866-mediated cell killing. Autophagy-modulating approaches could be a new way to enhance the antitumor activity of APO866 and related agents.
Collapse
Affiliation(s)
- Vanessa Ginet
- Department of Fundamental Neurosciences; Faculty of Biology and Medicine; University of Lausanne; Lausanne, Switzerland
| | - Julien Puyal
- Department of Fundamental Neurosciences; Faculty of Biology and Medicine; University of Lausanne; Lausanne, Switzerland
| | - Coralie Rummel
- Department of Fundamental Neurosciences; Faculty of Biology and Medicine; University of Lausanne; Lausanne, Switzerland
| | - Dominique Aubry
- Service and Central Laboratory of Hematology; University Hospital of Lausanne; Lausanne, Switzerland
| | - Caroline Breton
- Service and Central Laboratory of Hematology; University Hospital of Lausanne; Lausanne, Switzerland
| | - Anne-Julie Cloux
- Service and Central Laboratory of Hematology; University Hospital of Lausanne; Lausanne, Switzerland
| | - Somi R Majjigapu
- Laboratory of Glycochemistry and Asymmetric Synthesis; Swiss Federal Institute of Technology (EPFL); Batochime, Lausanne, Switzerland
| | - Bernard Sordat
- Laboratory of Glycochemistry and Asymmetric Synthesis; Swiss Federal Institute of Technology (EPFL); Batochime, Lausanne, Switzerland
| | - Pierre Vogel
- Laboratory of Glycochemistry and Asymmetric Synthesis; Swiss Federal Institute of Technology (EPFL); Batochime, Lausanne, Switzerland
| | - Santina Bruzzone
- Department of Experimental Medicine; Section of Biochemistry; University of Genoa; Genoa, Italy
| | - Alessio Nencioni
- Department of Internal Medicine; University of Genoa; Genoa, Italy
| | - Michel A Duchosal
- Service and Central Laboratory of Hematology; University Hospital of Lausanne; Lausanne, Switzerland
| | - Aimable Nahimana
- Service and Central Laboratory of Hematology; University Hospital of Lausanne; Lausanne, Switzerland
| |
Collapse
|
42
|
Ahmed FE. Development of novel diagnostic and prognostic molecular markers for sporadic colon cancer. Expert Rev Mol Diagn 2014; 5:337-52. [PMID: 15934812 DOI: 10.1586/14737159.5.3.337] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Gene expression studies are informative about changes in colon cancer, increase understanding of the biology of tumorigenesis and aid in developing diagnostic and prognostic markers. In this review, expression techniques used to examine the multistage process of colon cancer are discussed. Many genes have been found to differ in expression between normal and tumorigenic states, as early as the seemingly normal colonic crypts. The clinical usefulness of markers varies with stage, ethnicity and anatomic location of colon cancer. Thus, combinations of markers can be used to develop an approach to molecularly screen and follow the progression of this prevalent cancer.
Collapse
Affiliation(s)
- Farid E Ahmed
- The Brody School of Medicine at East Carolina University, Department of Radiation Oncology, Leo W. Jenkins Cancer Center, Greenville, NC 27858, USA.
| |
Collapse
|
43
|
Zerp SF, Vens C, Floot B, Verheij M, van Triest B. NAD⁺ depletion by APO866 in combination with radiation in a prostate cancer model, results from an in vitro and in vivo study. Radiother Oncol 2014; 110:348-54. [PMID: 24412016 DOI: 10.1016/j.radonc.2013.10.039] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 10/09/2013] [Accepted: 10/26/2013] [Indexed: 01/20/2023]
Abstract
BACKGROUND APO866 is a highly specific inhibitor of nicotinamide phosphoribosyltransferase (NAMPT), inhibition of which reduces cellular NAD(+) levels. In this study we addressed the potential of NAD(+) depletion as an anti-cancer strategy and assessed the combination with radiation. METHODS The anticipated radiosensitizing property of APO866 was investigated in prostate cancer cell lines PC3 and LNCaP in vitro and in PC3 xenografts in vivo. RESULTS We show that APO866 treatment leads to NAD(+) depletion. Combination experiments with radiation lead to a substantial decrease in clonogenic cell survival in PC3 and LNCaP cells. In PC3 xenografts, treatment with APO866 resulted in reduced intratumoral NAD(+) levels and induced significant tumor growth delay. Combined treatment of APO866 and fractionated radiation was more effective than the single modalities. Compared with untreated tumors, APO866 and radiation alone resulted in tumor growth delays of 14 days and 33 days, respectively, whereas the combination showed a significantly increased tumor growth delay of 65 days. CONCLUSIONS Our studies show that APO866-induced NAD(+) depletion enhances radiation responses in tumor cell survival in prostate cancer. However, the in vitro data do not reveal a solid cellular mechanism to exploit further clinical development at this moment.
Collapse
Affiliation(s)
- Shuraila F Zerp
- Division of Biological Stress Response, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Conchita Vens
- Division of Biological Stress Response, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Ben Floot
- Division of Biological Stress Response, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Marcel Verheij
- Division of Biological Stress Response, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands; Department of Radiation Oncology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands.
| | - Baukelien van Triest
- Department of Radiation Oncology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| |
Collapse
|
44
|
Pérez-Hernández AI, Catalán V, Gómez-Ambrosi J, Rodríguez A, Frühbeck G. Mechanisms linking excess adiposity and carcinogenesis promotion. Front Endocrinol (Lausanne) 2014; 5:65. [PMID: 24829560 PMCID: PMC4013474 DOI: 10.3389/fendo.2014.00065] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 04/15/2014] [Indexed: 12/17/2022] Open
Abstract
Obesity constitutes one of the most important metabolic diseases being associated to insulin resistance development and increased cardiovascular risk. Association between obesity and cancer has also been well established for several tumor types, such as breast cancer in post-menopausal women, colorectal, and prostate cancer. Cancer is the first death cause in developed countries and the second one in developing countries, with high incidence rates around the world. Furthermore, it has been estimated that 15-20% of all cancer deaths may be attributable to obesity. Tumor growth is regulated by interactions between tumor cells and their tissue microenvironment. In this sense, obesity may lead to cancer development through dysfunctional adipose tissue and altered signaling pathways. In this review, three main pathways relating obesity and cancer development are examined: (i) inflammatory changes leading to macrophage polarization and altered adipokine profile; (ii) insulin resistance development; and (iii) adipose tissue hypoxia. Since obesity and cancer present a high prevalence, the association between these conditions is of great public health significance and studies showing mechanisms by which obesity lead to cancer development and progression are needed to improve prevention and management of these diseases.
Collapse
Affiliation(s)
| | - Victoria Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
- *Correspondence: Gema Frühbeck, Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Avda. Pío XII 36, Pamplona 31008, Spain e-mail:
| |
Collapse
|
45
|
Genetic variants in NAMPT predict bladder cancer risk and prognosis in individuals from southwest Chinese Han group. Tumour Biol 2013; 35:4031-40. [PMID: 24363084 DOI: 10.1007/s13277-013-1527-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 12/06/2013] [Indexed: 12/27/2022] Open
Abstract
Nicotinamide phosphoribosyltransferase (Nampt) was served as a useful biomarker for tumorigenesis and for the prediction of cancer survival. In the present study, we analyzed the SNPs of the NAMPT gene and their impact on the susceptibility and prognosis for patients with bladder cancer (BC). The rs61330082, rs2505568 and rs9034 were selected and genotyped by polymerase chain reaction (PCR)-restriction fragment length polymorphism (RFLP) method in 407 patients with bladder cancer and 316 ethnicity-matched healthy control subjects. The genotyping method was confirmed by the DNA sequencing analysis. Statistically significant increased bladder cancer risk was found to be associated with the C allele and CC genotype of rs61330082; nevertheless, decreased bladder cancer risk was revealed to be associated with A allele and AT genotype of rs2505568. Stratified analyses revealed the rs61330082 to be statistically associated with increased bladder cancer risk in smokers and increased invasiveness of bladder cancer. The AT heterozygote of rs2505568 may prevent the recurrence of bladder cancer. Kaplan-Meier curves revealed a statistically significant association of rs2505568 with recurrence-free survival for total bladder cancer patients and non-muscle-invasive bladder cancer patients, and a statistically significant association of rs9034 with recurrence-free survival for muscle-invasive bladder cancer patients. Multiple Cox regression analysis identified the rs2505568 as a possible independent prognostic factor for recurrence-free survival in total bladder cancer patients. Our results suggested an important role for NAMPT in the pathogenesis of bladder cancer and SNPs of NAMPT gene might be a novel genetic biomarker for the prognosis of bladder cancer.
Collapse
|
46
|
Christensen MK, Erichsen KD, Olesen UH, Tjørnelund J, Fristrup P, Thougaard A, Nielsen SJ, Sehested M, Jensen PB, Loza E, Kalvinsh I, Garten A, Kiess W, Björkling F. Nicotinamide phosphoribosyltransferase inhibitors, design, preparation, and structure-activity relationship. J Med Chem 2013; 56:9071-88. [PMID: 24164086 DOI: 10.1021/jm4009949] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Existing pharmacological inhibitors for nicotinamide phosphoribosyltransferase (NAMPT) are promising therapeutics for treating cancer. By using medicinal and computational chemistry methods, the structure-activity relationship for novel classes of NAMPT inhibitors is described, and the compounds are optimized. Compounds are designed inspired by the NAMPT inhibitor APO866 and cyanoguanidine inhibitor scaffolds. In comparison with recently published derivatives, the new analogues exhibit an equally potent antiproliferative activity in vitro and comparable activity in vivo. The best performing compounds from these series showed subnanomolar antiproliferative activity toward a series of cancer cell lines (compound 15: IC50 0.025 and 0.33 nM, in A2780 (ovarian carcinoma) and MCF-7 (breast), respectively) and potent antitumor in vivo activity in well-tolerated doses in a xenograft model. In an A2780 xenograft mouse model with large tumors (500 mm(3)), compound 15 reduced the tumor volume to one-fifth of the starting volume at a dose of 3 mg/kg administered ip, bid, days 1-9. Thus, compounds found in this study compared favorably with compounds already in the clinic and warrant further investigation as promising lead molecules for the inhibition of NAMPT.
Collapse
|
47
|
Chen M, Wang Y, Li Y, Zhao L, Ye S, Wang S, Yu C, Xie H. Association of plasma visfatin with risk of colorectal cancer: An observational study of Chinese patients. Asia Pac J Clin Oncol 2013; 12:e65-74. [PMID: 23910020 DOI: 10.1111/ajco.12090] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
AIM To investigate the association between plasma visfatin levels and risk of early and advanced colorectal cancer (CRC). METHODS In total, 358 CRC patients and 286 controls were enrolled. According to the T factor of the TNM system. cancer patients were divided into two subgroups: early and advanced cancer. Levels of visfatin, anthropometric and metabolic parameters, which were classified as low, medium, and high, based on the tertile distributions in the control group, were determined. RESULTS The visfatin levels in patients with advanced and early cancer were higher than in controls (least significant difference test, P = 0.004 and 0.013, respectively). The patients in the highest tertile of visfatin concentration presented significantly higher odds for early and advanced CRC, adjusted for potential confounding factors (odds ratio 3.37; 95% CI, 1.93-8.37; P = 0.011; odds ratio 2.38; 95% CI: 1.82-8.35; P = 0.015, respectively). The visfatin level correlated significantly with waist:hip ratio (P < 0.05 for all) among case and control participants. Plasma visfatin levels in early and advanced CRC yielded a receiver operating characteristic curve area of 72 and 86%, respectively. The optimal sensitivity and specificity were 73% and 57% in discriminating between early CRC and normal controls while they were 76% and 68% in discriminating between advanced CRC and normal controls. CONCLUSION An increased level of visfatin was a strong risk factor for both early and advanced CRC in Chinese patients. Plasma visfatin levels might be a potential biomarker for CRC detection.
Collapse
Affiliation(s)
- Mingwei Chen
- Department of Endocrinology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Youming Wang
- Department of Endocrinology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yongxiang Li
- Division of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Lili Zhao
- Department of Endocrinology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Shuai Ye
- Department of Endocrinology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Shenyi Wang
- Division of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Changjun Yu
- Division of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Huijuan Xie
- Division of Endoscopy, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
48
|
Zhang C, Tong J, Huang G. Nicotinamide phosphoribosyl transferase (Nampt) is a target of microRNA-26b in colorectal cancer cells. PLoS One 2013; 8:e69963. [PMID: 23922874 PMCID: PMC3726743 DOI: 10.1371/journal.pone.0069963] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 06/13/2013] [Indexed: 12/22/2022] Open
Abstract
A number of cancers show increased expression of Nicotinamide phosphoribosyl transferase (Nampt). However, the mechanism through which Nampt is upregulated is unclear. In our study, we found that the Nampt-specific chemical inhibitor FK866 significantly inhibited cell survival and reduced nicotinamide adenine dinucleotide (NAD) levels in LoVo and SW480 cell lines. Bioinformatics analyses suggested that miR-26b targets Nampt mRNA. We identified Nampt as a new target of miR-26b and demonstrated that miR-26b inhibits Nampt expression at the protein and mRNA levels by binding to the Nampt 3′-UTR. Moreover, we found that miR-26b was down regulated in cancer tissues relative to that in adjacent normal tissues in 18 colorectal cancer patients. A statistically significant inverse correlation between miR-26b and Nampt expression was observed in samples from colorectal cancer patients and in 5 colorectal cell lines (HT-29, SW480, SW1116, LoVo, and HCT116). In addition, over expression of miR-26b strongly inhibited LoVo cell survival and invasion, an effect partially abrogated by the addition of NAD. In conclusion, this study demonstrated that the NAD-salvaging biosynthesis pathway involving Nampt might play a role in colorectal cancer cell survival. MiR-26b may serve as a tumor suppressor by targeting Nampt.
Collapse
Affiliation(s)
- Chenpeng Zhang
- Department of Nuclear Medicine, Ren Ji hospital, School of medicine, Shanghai Jiao Tong University, Shanghai, China
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Jinlu Tong
- Department of Gastroenterology, Shanghai Institute of Gastrointestinal Diseases; Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Gang Huang
- Department of Nuclear Medicine, Ren Ji hospital, School of medicine, Shanghai Jiao Tong University, Shanghai, China
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
- * E-mail: .
| |
Collapse
|
49
|
Tian W, Zhu Y, Wang Y, Teng F, Zhang H, Liu G, Ma X, Sun D, Rohan T, Xue F. Visfatin, a potential biomarker and prognostic factor for endometrial cancer. Gynecol Oncol 2013; 129:505-12. [DOI: 10.1016/j.ygyno.2013.02.022] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 02/14/2013] [Accepted: 02/15/2013] [Indexed: 12/21/2022]
|
50
|
Soulet F, Kilarski WW, Roux-Dalvai F, Herbert JMJ, Sacewicz I, Mouton-Barbosa E, Bicknell R, Lalor P, Monsarrat B, Bikfalvi A. Mapping the extracellular and membrane proteome associated with the vasculature and the stroma in the embryo. Mol Cell Proteomics 2013; 12:2293-312. [PMID: 23674615 DOI: 10.1074/mcp.m112.024075] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
In order to map the extracellular or membrane proteome associated with the vasculature and the stroma in an embryonic organism in vivo, we developed a biotinylation technique for chicken embryo and combined it with mass spectrometry and bioinformatic analysis. We also applied this procedure to implanted tumors growing on the chorioallantoic membrane or after the induction of granulation tissue. Membrane and extracellular matrix proteins were the most abundant components identified. Relative quantitative analysis revealed differential protein expression patterns in several tissues. Through a bioinformatic approach, we determined endothelial cell protein expression signatures, which allowed us to identify several proteins not yet reported to be associated with endothelial cells or the vasculature. This is the first study reported so far that applies in vivo biotinylation, in combination with robust label-free quantitative proteomics approaches and bioinformatic analysis, to an embryonic organism. It also provides the first description of the vascular and matrix proteome of the embryo that might constitute the starting point for further developments.
Collapse
|