1
|
Zhang Y, Chen H, You S, Chi X, Chen Y, Wu Z, Zheng J, Cheng X, Liu J, Li J. Preliminary investigation of nicotinamide N-methyltransferase as an HBV-specific biomarker for hepatocellular carcinoma diagnosis. Int J Biol Markers 2025:3936155251330664. [PMID: 40329575 DOI: 10.1177/03936155251330664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
BackgroundNicotinamide N-methyltransferase (NNMT), a metabolic enzyme in the liver, has been implicated in various biological processes, and its high expression in hepatocellular carcinoma has been linked to tumor metastasis and poor prognosis. However, its potential as a serum biomarker for hepatocellular carcinoma diagnosis remains unexplored.MethodsA total of 172 subjects were included in this study, consisting of 71 hepatocellular carcinoma patients (64 with hepatitis B virus (HBV)-associated hepatocellular carcinoma and 7 with non-HBV-associated hepatocellular carcinoma), as well as 70 healthy controls and 31 HBV-infected individuals. Serum NNMT levels were measured, and clinical-pathological correlations were analyzed. The diagnostic efficacy of serum NNMT for HBV-related hepatocellular carcinoma was evaluated using receiver operating characteristic (ROC) curve analysis.ResultsSerum NNMT levels were significantly elevated in HBV-infected individuals and correlated with poorer prognosis, including reduced overall survival and shorter disease-free survival. Kaplan-Meier analysis revealed that low NNMT expression was associated with longer overall survival (75 vs. 12 months, P < 0.0001) and disease-free survival (21.5 vs. 5 months, P < 0.01). In HBV-related hepatocellular carcinoma patients, NNMT levels correlated with biochemical markers including alfa-fetoprotein, aspartate transaminase, triglycerides, total cholesterol, low-density lipoprotein, apolipoprotein B, TB, and albumin, with decreased albumin, and high-density lipoprotein levels promoting NNMT expression. ROC analysis showed that NNMT outperformed alfa-fetoprotein (area under the curve (AUC) 0.869 vs. 0.775), with a sensitivity of 95.2%, specificity of 87.9%, and a combined AUC of 0.947, demonstrating its superior diagnostic value for HBV-related hepatocellular carcinoma.ConclusionsSerum NNMT is a promising biomarker for predicting the risk of hepatocellular carcinoma in HBV-infected individuals and may serve as an indicator for the prognosis of hepatocellular carcinoma patients.
Collapse
Affiliation(s)
- Yi Zhang
- Vascular Surgery Department, Xiamen Hospital of Traditional Chinese Medicine, Xiamen, Fujian Province, China
| | - Huayang Chen
- Department of Colorectal Surgery, Xiamen Hospital of Traditional Chinese Medicine, Xiamen, Fujian Province, China
| | - Song You
- Department of Hepatobiliary Surgery, Meng Chao Hepatobiliary Hospital Affiliated to Fujian Medical University, Fuzhou, Fujian Province, China
| | - Xiaoqin Chi
- Department of Hepatobiliary Surgery, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Xiamen Translational Medical Key Laboratory of Digestive System Tumor, Zhongshan Hospital of Xiamen University, Fujian Province, China
| | - Yongxi Chen
- Vascular Surgery Department, Xiamen Hospital of Traditional Chinese Medicine, Xiamen, Fujian Province, China
| | - Zhiyong Wu
- Vascular Surgery Department, Xiamen Hospital of Traditional Chinese Medicine, Xiamen, Fujian Province, China
| | - Jiepeng Zheng
- Vascular Surgery Department, Xiamen Hospital of Traditional Chinese Medicine, Xiamen, Fujian Province, China
| | - Xin Cheng
- Vascular Surgery Department, Xiamen Hospital of Traditional Chinese Medicine, Xiamen, Fujian Province, China
| | - Jianming Liu
- Department of Hepatobiliary Surgery, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Xiamen Translational Medical Key Laboratory of Digestive System Tumor, Zhongshan Hospital of Xiamen University, Fujian Province, China
| | - Jie Li
- Department of Hepatobiliary Surgery, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Xiamen Translational Medical Key Laboratory of Digestive System Tumor, Zhongshan Hospital of Xiamen University, Fujian Province, China
| |
Collapse
|
2
|
Park J, Shin EJ, Kim TH, Yang JH, Ki SH, Kang KW, Kim KM. Exploring NNMT: from metabolic pathways to therapeutic targets. Arch Pharm Res 2024; 47:893-913. [PMID: 39604638 DOI: 10.1007/s12272-024-01519-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024]
Abstract
Cellular metabolism-related epigenetic modulation plays a pivotal role in the maintenance of cellular homeostasis. Nicotinamide N-methyltransferase (NNMT) serves as a crucial link between cellular metabolism and epigenetics by catalyzing nicotinamide methylation using the universal methyl donor S-adenosyl-L-methionine. This direct connection bridges the methylation-mediated one-carbon metabolism with nicotinamide adenine dinucleotide levels. Numerous studies have revealed tissue-specific differences in NNMT expression and activity, indicating that its varied physiological and pathological roles depend on its distribution. In this review, we provide an overview of the NNMT involvement in various pathological conditions, including cancer, liver disease, obesity, diabetes, brain disease, pulmonary disease, cardiovascular disease, and kidney disease. By synthesizing this information, our article aims to enhance our understanding of the cellular mechanisms underlying NNMT biology related to diverse diseases and lay the molecular groundwork for developing therapeutic strategies for pharmacological interventions.
Collapse
Affiliation(s)
- Jeongwoo Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
- Department of Biomedical Science, College of Natural Science, Chosun University, Gwangju, 61452, Republic of Korea
- Institute of Well-Aging Medicare & Chosun University G-LAMP Project Group, Chosun University, Gwangju, 61452, Republic of Korea
| | - Eun Jin Shin
- Department of Biomedical Science, College of Natural Science, Chosun University, Gwangju, 61452, Republic of Korea
- Institute of Well-Aging Medicare & Chosun University G-LAMP Project Group, Chosun University, Gwangju, 61452, Republic of Korea
- Department of Integrative Biological Sciences & BK21 FOUR Educational Research Group for Age-Associated Disorder Control Technology, Chosun University, Gwangju, 61452, Republic of Korea
| | - Tae Hyun Kim
- Drug Information Research Institute, Muscle Physiome Research Center, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, South Korea
| | - Ji Hye Yang
- College of Korean Medicine, Dongshin University, Naju, Jeollanam-Do, 58245, Republic of Korea
| | - Sung Hwan Ki
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea
| | - Keon Wook Kang
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Kyu Min Kim
- Department of Biomedical Science, College of Natural Science, Chosun University, Gwangju, 61452, Republic of Korea.
- Institute of Well-Aging Medicare & Chosun University G-LAMP Project Group, Chosun University, Gwangju, 61452, Republic of Korea.
- Department of Integrative Biological Sciences & BK21 FOUR Educational Research Group for Age-Associated Disorder Control Technology, Chosun University, Gwangju, 61452, Republic of Korea.
| |
Collapse
|
3
|
Pozzi V, Molinelli E, Campagna R, Serritelli EN, Cecati M, De Simoni E, Sartini D, Goteri G, Martin NI, van Haren MJ, Salvolini E, Simonetti O, Offidani A, Emanuelli M. Knockdown of nicotinamide N-methyltransferase suppresses proliferation, migration, and chemoresistance of Merkel cell carcinoma cells in vitro. Hum Cell 2024; 37:729-738. [PMID: 38504052 PMCID: PMC11016511 DOI: 10.1007/s13577-024-01047-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 02/21/2024] [Indexed: 03/21/2024]
Abstract
Merkel cell carcinoma (MCC) is an aggressive skin cancer, with a propensity for early metastasis. Therefore, early diagnosis and the identification of novel targets become fundamental. The enzyme nicotinamide N-methyltransferase (NNMT) catalyzes the reaction of N-methylation of nicotinamide and other analogous compounds. Although NNMT overexpression was reported in many malignancies, the significance of its dysregulation in cancer cell phenotype was partly clarified. Several works demonstrated that NNMT promotes cancer cell proliferation, migration, and chemoresistance. In this study, we investigated the possible involvement of this enzyme in MCC. Preliminary immunohistochemical analyses were performed to evaluate NNMT expression in MCC tissue specimens. To explore the enzyme function in tumor cell metabolism, MCC cell lines have been transfected with plasmids encoding for short hairpin RNAs (shRNAs) targeting NNMT mRNA. Preliminary immunohistochemical analyses showed elevated NNMT expression in MCC tissue specimens. The effect of enzyme downregulation on cell proliferation, migration, and chemosensitivity was then evaluated through MTT, trypan blue, and wound healing assays. Data obtained clearly demonstrated that NNMT knockdown is associated with a decrease of cell proliferation, viability, and migration, as well as with enhanced sensitivity to treatment with chemotherapeutic drugs. Taken together, these results suggest that NNMT could represent an interesting MCC biomarker and a promising target for targeted anti-cancer therapy.
Collapse
Affiliation(s)
- Valentina Pozzi
- Department of Clinical Sciences, Polytechnic University of Marche, 60020, Ancona, Italy
| | - Elisa Molinelli
- Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60020, Ancona, Italy
| | - Roberto Campagna
- Department of Clinical Sciences, Polytechnic University of Marche, 60020, Ancona, Italy.
| | - Emma N Serritelli
- Department of Clinical Sciences, Polytechnic University of Marche, 60020, Ancona, Italy
| | - Monia Cecati
- Department of Clinical Sciences, Polytechnic University of Marche, 60020, Ancona, Italy
| | - Edoardo De Simoni
- Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60020, Ancona, Italy
| | - Davide Sartini
- Department of Clinical Sciences, Polytechnic University of Marche, 60020, Ancona, Italy.
| | - Gaia Goteri
- Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, 60020, Ancona, Italy
| | - Nathaniel I Martin
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, Sylviusweg 72, 2333 BE, Leiden, The Netherlands
| | - Matthijs J van Haren
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, Sylviusweg 72, 2333 BE, Leiden, The Netherlands
| | - Eleonora Salvolini
- Department of Clinical Sciences, Polytechnic University of Marche, 60020, Ancona, Italy
| | - Oriana Simonetti
- Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60020, Ancona, Italy
| | - Annamaria Offidani
- Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60020, Ancona, Italy
| | - Monica Emanuelli
- Department of Clinical Sciences, Polytechnic University of Marche, 60020, Ancona, Italy
- New York-Marche Structural Biology Center (NY-MaSBiC), Polytechnic University of Marche, 60131, Ancona, Italy
| |
Collapse
|
4
|
Vellky JE, Wu Y, Moline D, Drnevich J, Maienschein-Cline M, Valyi-Nagy K, Kajdacsy-Balla A, Vander Griend DJ. Single-cell RNA sequencing of human prostate basal epithelial cells reveals zone-specific cellular populations and gene expression signatures. J Pathol 2024; 262:212-225. [PMID: 37984408 PMCID: PMC10842206 DOI: 10.1002/path.6227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 08/24/2023] [Accepted: 10/05/2023] [Indexed: 11/22/2023]
Abstract
Despite evidence of genetic signatures in normal tissue correlating with disease risk, prospectively identifying genetic drivers and cell types that underlie subsequent pathologies has historically been challenging. The human prostate is an ideal model to investigate this phenomenon because it is anatomically segregated into three glandular zones (central, peripheral, and transition) that develop differential pathologies: prostate cancer in the peripheral zone (PZ) and benign prostatic hyperplasia (BPH) in the transition zone (TZ), with the central zone (CZ) rarely developing disease. More specifically, prostatic basal cells have been implicated in differentiation and proliferation during prostate development and regeneration; however, the contribution of zonal variation and the critical role of basal cells in prostatic disease etiology are not well understood. Using single-cell RNA sequencing of primary prostate epithelial cultures, we elucidated organ-specific, zone-specific, and cluster-specific gene expression differences in basal cells isolated from human prostate and seminal vesicle (SV). Aggregated analysis identified ten distinct basal clusters by Uniform Manifold Approximation and Projection. Organ specificity compared gene expression in SV with the prostate. As expected, SV cells were distinct from prostate cells by clustering, gene expression, and pathway analysis. For prostate zone specificity, we identified two CZ-specific clusters, while the TZ and PZ populations clustered together. Despite these similarities, differential gene expression was identified between PZ and TZ samples that correlated with gene expression profiles in prostate cancer and BPH, respectively. Zone-specific profiles and cell type-specific markers were validated using immunostaining and bioinformatic analyses of publicly available RNA-seq datasets. Understanding the baseline differences at the organ, zonal, and cellular level provides important insight into the potential drivers of prostatic disease and guides the investigation of novel preventive or curative treatments. Importantly, this study identifies multiple prostate basal cell populations and cell type-specific gene signatures within prostate basal epithelial cells that have potential critical roles in driving prostatic diseases. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Jordan E Vellky
- Department of Pathology, The University of Illinois at Chicago, Chicago, IL, USA
| | - Yaqi Wu
- Department of Pathology, The University of Illinois at Chicago, Chicago, IL, USA
| | - Daniel Moline
- Committee on Development, Regeneration, and Stem Cell Biology (DRSB), The University of Chicago, Chicago, IL, USA
| | - Jenny Drnevich
- High Performance Computing in Biology (HPCBio); Biotechnology Center; Roy J. Carver Biotechnology Center, The University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | | | - Klara Valyi-Nagy
- Department of Pathology, The University of Illinois at Chicago, Chicago, IL, USA
| | - Andre Kajdacsy-Balla
- Department of Pathology, The University of Illinois at Chicago, Chicago, IL, USA
| | | |
Collapse
|
5
|
Masci D, Naro C, Puxeddu M, Urbani A, Sette C, La Regina G, Silvestri R. Recent Advances in Drug Discovery for Triple-Negative Breast Cancer Treatment. Molecules 2023; 28:7513. [PMID: 38005235 PMCID: PMC10672974 DOI: 10.3390/molecules28227513] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/02/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is one of the most heterogeneous and aggressive breast cancer subtypes with a high risk of death on recurrence. To date, TNBC is very difficult to treat due to the lack of an effective targeted therapy. However, recent advances in the molecular characterization of TNBC are encouraging the development of novel drugs and therapeutic combinations for its therapeutic management. In the present review, we will provide an overview of the currently available standard therapies and new emerging therapeutic strategies against TNBC, highlighting the promises that newly developed small molecules, repositioned drugs, and combination therapies have of improving treatment efficacy against these tumors.
Collapse
Affiliation(s)
- Domiziana Masci
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168 Rome, Italy; (D.M.); (A.U.)
| | - Chiara Naro
- Department of Neurosciences, Section of Human Anatomy, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168 Rome, Italy; (C.N.); (C.S.)
- GSTeP-Organoids Research Core Facility, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Michela Puxeddu
- Laboratory Affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (M.P.); (G.L.R.)
| | - Andrea Urbani
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168 Rome, Italy; (D.M.); (A.U.)
| | - Claudio Sette
- Department of Neurosciences, Section of Human Anatomy, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168 Rome, Italy; (C.N.); (C.S.)
- GSTeP-Organoids Research Core Facility, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Giuseppe La Regina
- Laboratory Affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (M.P.); (G.L.R.)
| | - Romano Silvestri
- Laboratory Affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (M.P.); (G.L.R.)
| |
Collapse
|
6
|
Perez MF, Sarkies P. Histone methyltransferase activity affects metabolism in human cells independently of transcriptional regulation. PLoS Biol 2023; 21:e3002354. [PMID: 37883365 PMCID: PMC10602318 DOI: 10.1371/journal.pbio.3002354] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 09/27/2023] [Indexed: 10/28/2023] Open
Abstract
The N-terminal tails of eukaryotic histones are frequently posttranslationally modified. The role of these modifications in transcriptional regulation is well-documented. However, the extent to which the enzymatic processes of histone posttranslational modification might affect metabolic regulation is less clear. Here, we investigated how histone methylation might affect metabolism using metabolomics, proteomics, and RNA-seq data from cancer cell lines, primary tumour samples and healthy tissue samples. In cancer, the expression of histone methyltransferases (HMTs) was inversely correlated to the activity of NNMT, an enzyme previously characterised as a methyl sink that disposes of excess methyl groups carried by the universal methyl donor S-adenosyl methionine (SAM or AdoMet). In healthy tissues, histone methylation was inversely correlated to the levels of an alternative methyl sink, PEMT. These associations affected the levels of multiple histone marks on chromatin genome-wide but had no detectable impact on transcriptional regulation. We show that HMTs with a variety of different associations to transcription are co-regulated by the Retinoblastoma (Rb) tumour suppressor in human cells. Rb-mutant cancers show increased total HMT activity and down-regulation of NNMT. Together, our results suggest that the total activity of HMTs affects SAM metabolism, independent of transcriptional regulation.
Collapse
Affiliation(s)
- Marcos Francisco Perez
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
- Department of Cells and Tissues, Instituto de Biologia Molecular de Barcelona (IBMB), CSIC, Barcelona, Spain
| | - Peter Sarkies
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
7
|
Wang Y, Zhou X, Lei Y, Chu Y, Yu X, Tong Q, Zhu T, Yu H, Fang S, Li G, Wang L, Wang GY, Xie X, Zhang J. NNMT contributes to high metastasis of triple negative breast cancer by enhancing PP2A/MEK/ERK/c-Jun/ABCA1 pathway mediated membrane fluidity. Cancer Lett 2022; 547:215884. [PMID: 35988817 DOI: 10.1016/j.canlet.2022.215884] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 08/12/2022] [Accepted: 08/12/2022] [Indexed: 11/02/2022]
Abstract
Elucidating the mechanism for high metastasis capacity of triple negative breast cancers (TNBC) is crucial to improve treatment outcomes of TNBC. We have recently reported that nicotinamide N-methyltransferase (NNMT) is overexpressed in breast cancer, especially in TNBC, and predicts poor survival of patients undergoing chemotherapy. Here, we aimed to determine the function and mechanism of NNMT on metastasis of TNBC. Additionally, analysis of public datasets indicated that NNMT is involved in cholesterol metabolism. In vitro, NNMT overexpression promoted migration and invasion of TNBCs by reducing cholesterol levels in the cytoplasm and cell membrane. Mechanistically, NNMT activated MEK/ERK/c-Jun/ABCA1 pathway by repressing protein phosphatase 2A (PP2A) activity leading to cholesterol efflux and membrane fluidity enhancement, thereby promoting the epithelial-mesenchymal transition (EMT) of TNBCs. In vivo, the metastasis capacity of TNBCs was weakened by targeting NNMT. Collectively, our findings suggest a new molecular mechanism involving NNMT in metastasis and poor survival of TNBC mediated by PP2A and affecting cholesterol metabolism.
Collapse
Affiliation(s)
- Yanzhong Wang
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Department of Clinical Laboratory, Xiasha Campus, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, Zhejiang, PR China
| | - Xi Zhou
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, Zhejiang, PR China
| | - Yinjiao Lei
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, Zhejiang, PR China
| | - Yadong Chu
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, Zhejiang, PR China; Department of Clinical Laboratory, Zhejiang Armed Police Corps Hospital, Hangzhou, Zhejiang, PR China
| | - Xingtong Yu
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, Zhejiang, PR China
| | - Qingchao Tong
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, Zhejiang, PR China
| | - Tao Zhu
- Department of Pathology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China
| | - Haitao Yu
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, Zhejiang, PR China
| | - Sining Fang
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, Zhejiang, PR China
| | - Guoli Li
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, Zhejiang, PR China
| | - Linbo Wang
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China
| | - Gavin Y Wang
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, 29425, SC, USA; Cancer Cell Biology Program of the Hollings Cancer Center, Medical University of South Carolina, Charleston, 29425, SC, USA
| | - Xinyou Xie
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Department of Clinical Laboratory, Xiasha Campus, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, Zhejiang, PR China.
| | - Jun Zhang
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Department of Clinical Laboratory, Xiasha Campus, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, Zhejiang, PR China.
| |
Collapse
|
8
|
Pozzi V, Campagna R, Sartini D, Emanuelli M. Nicotinamide N-Methyltransferase as Promising Tool for Management of Gastrointestinal Neoplasms. Biomolecules 2022; 12:biom12091173. [PMID: 36139012 PMCID: PMC9496617 DOI: 10.3390/biom12091173] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/21/2022] [Accepted: 08/23/2022] [Indexed: 12/24/2022] Open
Abstract
Gastrointestinal (GI) neoplasms include esophageal, gastric, colorectal, hepatic, and pancreatic cancers. They are characterized by asymptomatic behavior, being responsible for diagnostic delay. Substantial refractoriness to chemo- and radiotherapy, exhibited by late-stage tumors, contribute to determine poor patient outcome. Therefore, it is of outmost importance to identify new molecular targets for the development of effective therapeutic strategies. In this study, we focused on the enzyme nicotinamide N-methyltransferase (NNMT), which catalyzes the N-methylation reaction of nicotinamide and whose overexpression has been reported in numerous neoplasms, including GI cancers. The aim of this review was to report data illustrating NNMT involvement in these tumors, highlighting its contribution to tumor cell phenotype. Cited works clearly demonstrate the interesting potential use of enzyme level determination for both diagnostic and prognostic purposes. NNMT was also found to positively affect cell viability, proliferation, migration, and invasiveness, contributing to sustain in vitro and in vivo tumor growth and metastatic spread. Moreover, enzyme upregulation featuring tumor cells was significantly associated with enhancement of resistance to treatment with chemotherapeutic drugs. Taken together, these results strongly suggest the possibility to target NNMT for setup of molecular-based strategies to effectively treat GI cancers.
Collapse
Affiliation(s)
- Valentina Pozzi
- Department of Clinical Sciences, Polytechnic University of Marche, 60020 Ancona, Italy
| | - Roberto Campagna
- Department of Clinical Sciences, Polytechnic University of Marche, 60020 Ancona, Italy
| | - Davide Sartini
- Department of Clinical Sciences, Polytechnic University of Marche, 60020 Ancona, Italy
- Correspondence: ; Tel.: +39-071-2204673
| | - Monica Emanuelli
- Department of Clinical Sciences, Polytechnic University of Marche, 60020 Ancona, Italy
- New York-Marche Structural Biology Center (NY-MaSBiC), Polytechnic University of Marche, 60131 Ancona, Italy
| |
Collapse
|
9
|
Çelik ZE, Akar S, Fındık S, Aytekin E, Çelik Ç. Nicotinamide N-methyltransferase expression in squamous cell carcinoma of the vulva. J OBSTET GYNAECOL 2022; 42:2325-2330. [PMID: 35499473 DOI: 10.1080/01443615.2022.2049722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The molecular pathways involved in the development of vulvar squamous cell carcinoma (SCC) cancer are not completely known. Nicotinamide N-methyltransferase (NNMT) is a cytosolic enzyme associated with tumorigenesis and metastasis in a variety of cancers. Its role in vulvar cancer has not been studied, previously. Vulvar SCC, high and low grade squamous intraepithelial lesions (SILs) and benign squamous hyperplasia were analysed immunohistochemically. The mean staining score for vulvar SCC was significantly higher than the score for vulvar squamous hyperplasia (p<.001). The mean relapse-free survival for patients with low and high NNMT expression was 41.4 months (95% CI: 25.6-57.2) and 19.8 months (95% CI: 3.0-36.6), respectively (p=.035). The mean disease-specific survival for patients with low and high NNMT expression was 75.8 months (95% CI: 57.5-94.2) and 27.8 months (95% CI 12.2-43.4), respectively (p=.015). Although quite preliminary, this study showed that NNMT expression was elevated in vulvar SCC compared to benign and premalignant lesions. Additionally, elevated NNMT expression was associated with poor survival. Impact StatementWhat is already known on this subject? Nicotinamide N-methyltransferase (NNMT) is a methyltransferase, associated with tumour progression, spread and poor prognosis in a variety of cancers. Its upregulation can lead to DNA hypomethylation, which can in turn result in the activation of proto-oncogenes and deactivation of tumour suppressor genes.What do the results of this study add? Although quite preliminary, this study showed that NNMT expression was elevated in vulvar SCC compared to benign and premalignant lesions. Additionally, elevated NNMT expression was associated with poor survival.What are the implications of these findings for clinical practice and/or further research? NNMT has been regarded as a potential target of cancer therapy and its role in vulvar cancer has not been studied, previously. This is the first study to investigate the expression of NNMT in vulvar cancer and associate NNMT elevation with poor survival. NNMT can further be investigated as a possible target of vulvar cancer therapy.
Collapse
Affiliation(s)
| | - Serra Akar
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Selcuk University Hospital, Selcuk University Medical School, Konya, Turkey
| | - Sıddıka Fındık
- Department of Pathology, Meram Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Emine Aytekin
- Department of Obstetrics and Gynecology, Selcuk University Hospital, Selcuk University Medical School, Konya, Turkey
| | - Çetin Çelik
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Selcuk University Hospital, Selcuk University Medical School, Konya, Turkey
| |
Collapse
|
10
|
Tumor stromal nicotinamide N-methyltransferase overexpression as a prognostic biomarker for poor clinical outcome in early-stage colorectal cancer. Sci Rep 2022; 12:2767. [PMID: 35177765 PMCID: PMC8854702 DOI: 10.1038/s41598-022-06772-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/04/2022] [Indexed: 01/02/2023] Open
Abstract
In a quest for prognostic biomarkers in early-stage colorectal cancer, we investigated NNMT (nicotinamide N-methyltransferase) in large cohorts of patients. Immunohistochemical examination of 679 patients illustrates that NNMT protein is predominantly expressed in the cancer stroma at varying levels, and about 20% of cancer tissues overexpress NNMT when compared to levels observed in normal colorectal mucosa. Clinical correlation analyses of 572 patients with early-stage cancers reveal that NNMT protein overexpression is significantly associated with shorter overall and disease-free survival, but no such correlation is found in late-stage colorectal cancer. Analyses of TCGA and CPTAC colorectal cancer cohorts show that NNMT mRNA expression is positively correlated with protein levels, is significantly higher in CIMP-high or MSI subtypes than in CIMP-low or MSS subtypes, and is positively correlated with its paralog INMT but not with its interaction partners such as PNMT, ADK, APP, ATF6, BMF, BRD4, CDC37, or CRYZ. In early-stage cancers, NNMT expression is higher in BRAF-mutated than in BRAF wild type tumors but is not affected by KRAS or PIK3CA mutation status. As a cancer stromal protein with important roles in metabolism and cancer epigenetics, NNMT is emerging as a promising biomarker for risk stratification of early-stage cancers.
Collapse
|
11
|
The Utility of Nicotinamide N-Methyltransferase as a Potential Biomarker to Predict the Oncological Outcomes for Urological Cancers: An Update. Biomolecules 2021; 11:biom11081214. [PMID: 34439880 PMCID: PMC8393883 DOI: 10.3390/biom11081214] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/06/2021] [Accepted: 08/09/2021] [Indexed: 01/03/2023] Open
Abstract
Nicotinamide N-methyltransferase (NNMT) catalyzes the N-methylation reaction of nicotinamide, using S-adenosyl-L-methionine as the methyl donor. Enzyme overexpression has been described in many non-neoplastic diseases, as well as in a wide range of solid malignancies. This review aims to report and discuss evidence available in scientific literature, dealing with NNMT expression and the potential involvement in main urologic neoplasms, namely, renal, bladder and prostate cancers. Data illustrated in the cited studies clearly demonstrated NNMT upregulation (pathological vs. normal tissue) in association with these aforementioned tumors. In addition to this, enzyme levels were also found to correlate with key prognostic parameters and patient survival. Interestingly, NNMT overexpression also emerged in peripheral body fluids, such as blood and urine, thus leading to candidate the enzyme as promising biomarker for the early and non-invasive detection of these cancers. Examined results undoubtedly showed NNMT as having the capacity to promote cell proliferation, migration and invasiveness, as well as its potential participation in fundamental events highlighting cancer progression, metastasis and resistance to chemo- and radiotherapy. In the light of this evidence, it is reasonable to attribute to NNMT a promising role as a potential biomarker for the diagnosis and prognosis of urologic neoplasms, as well as a molecular target for effective anti-cancer treatment.
Collapse
|
12
|
Thamim M, Thirumoorthy K. Computational studies of selective N-methylation in nicotinamide: Epigenetic reprogramming in cancer. COMPUT THEOR CHEM 2021. [DOI: 10.1016/j.comptc.2020.113058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
13
|
Buckup M, Rice MA, Hsu EC, Garcia-Marques F, Liu S, Aslan M, Bermudez A, Huang J, Pitteri SJ, Stoyanova T. Plectin is a regulator of prostate cancer growth and metastasis. Oncogene 2021; 40:663-676. [PMID: 33219316 PMCID: PMC8078627 DOI: 10.1038/s41388-020-01557-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 10/27/2020] [Accepted: 11/02/2020] [Indexed: 12/14/2022]
Abstract
Prostate cancer is responsible for over 30,000 US deaths annually, attributed largely to incurable metastatic disease. Here, we demonstrate that high levels of plectin are associated with localized and metastatic human prostate cancer when compared to benign prostate tissues. Knock-down of plectin inhibits prostate cancer cell growth and colony formation in vitro, and growth of prostate cancer xenografts in vivo. Plectin knock-down further impairs aggressive and invasive cellular behavior assessed by migration, invasion, and wound healing in vitro. Consistently, plectin knock-down cells have impaired metastatic colonization to distant sites including liver, lung, kidney, bone, and genitourinary system. Plectin knock-down inhibited number of metastases per organ, as well as decreased overall metastatic burden. To gain insights into the role of plectin in prostate cancer growth and metastasis, we performed proteomic analysis of prostate cancer plectin knock-down xenograft tissues. Gene set enrichment analysis shows an increase in levels of proteins involved with extracellular matrix and laminin interactions, and a decrease in levels of proteins regulating amino acid metabolism, cytoskeletal proteins, and cellular response to stress. Collectively these findings demonstrate that plectin is an important regulator of prostate cancer cell growth and metastasis.
Collapse
Affiliation(s)
- Mark Buckup
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Palo Alto, CA, USA
| | - Meghan A Rice
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Palo Alto, CA, USA
| | - En-Chi Hsu
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Palo Alto, CA, USA
| | - Fernando Garcia-Marques
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Palo Alto, CA, USA
| | - Shiqin Liu
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Palo Alto, CA, USA
| | - Merve Aslan
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Palo Alto, CA, USA
| | - Abel Bermudez
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Palo Alto, CA, USA
| | - Jiaoti Huang
- Department of Pathology, Duke University, Durham, NC, USA
| | - Sharon J Pitteri
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Palo Alto, CA, USA
| | - Tanya Stoyanova
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Palo Alto, CA, USA.
| |
Collapse
|
14
|
Cui Y, Yang D, Wang W, Zhang L, Liu H, Ma S, Guo W, Yao M, Zhang K, Li W, Zhang Y, Guan F. Nicotinamide N-methyltransferase decreases 5-fluorouracil sensitivity in human esophageal squamous cell carcinoma through metabolic reprogramming and promoting the Warburg effect. Mol Carcinog 2020; 59:940-954. [PMID: 32367570 DOI: 10.1002/mc.23209] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/20/2020] [Accepted: 04/23/2020] [Indexed: 12/28/2022]
Abstract
Esophageal squamous cell carcinoma (ESCC) is a common malignant tumor with poor prognosis. And different individuals respond to the same drug differently. Increasing evidence has confirmed that metabolism reprogramming was involved in the drug sensitivity of tumor cells. However, the potential molecular mechanism of 5-fluorouracil (5-FU) sensitivity remains to be elucidated in ESCC cells. In this study, we found that the 5-FU sensitivity of TE1 cells was lower than that of EC1 and Eca109 cells. Gas chromatography-mass spectrometry analysis results showed that nicotinate and nicotinamide metabolism and tricarboxylic acid cycle were significantly different in these three cell lines. Nicotinamide N-methyltransferase (NNMT), a key enzyme of nicotinate and nicotinamide metabolism, was significantly higher expressed in TE1 cells than that in EC1 and Eca109 cells. Therefore, the function of NNMT on 5-FU sensitivity was analyzed in vitro and in vivo. NNMT downregulation significantly increased 5-FU sensitivity in TE1 cells. Meanwhile, the glucose consumption and lactate production were decreased, and the expression of glycolysis-related enzymes hexokinase 2, lactate dehydrogenase A, and phosphoglycerate mutase 1 were downregulated in NNMT knockdown TE1 cells. Besides, overexpression of NNMT in EC1 and Eca109 cells caused the opposite effects. Moreover, when glycolysis was inhibited by 2-deoxyglucose, the roles of NNMT on 5-FU sensitivity was weakened. In vivo experiments showed that NNMT knockdown significantly increased the sensitivity of xenografts to 5-FU and suppressed the Warburg effect. Overall, these results demonstrated that NNMT decreases 5-FU sensitivity in human ESCC cells through promoting the Warburg effect, suggesting that NNMT may contribute to predict the treatment effects of the clinical chemotherapy in ESCC.
Collapse
Affiliation(s)
- Yanyan Cui
- School of Life Sciences, Zhengzhou University, Zhengzhou
| | - Dawei Yang
- Zhongyuan Academy of Biological Medicine, Liaocheng People's Hospital, Liaocheng, China
| | - Wenjie Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou
| | - Luyu Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou
| | - Hongtao Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou
| | - Shanshan Ma
- School of Life Sciences, Zhengzhou University, Zhengzhou
| | - Wenna Guo
- School of Life Sciences, Zhengzhou University, Zhengzhou
| | - Minghao Yao
- School of Life Sciences, Zhengzhou University, Zhengzhou
| | - Kun Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou
| | - Wencai Li
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanting Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou
| | - Fangxia Guan
- School of Life Sciences, Zhengzhou University, Zhengzhou.,Clinical Research Guidance Center, Henan Provincial People's Hospital, Zhengzhou, China
| |
Collapse
|
15
|
Expression and Clinical Significance of Nicotinamide N-Methyltransferase in Cervical Squamous Cell Carcinoma. Int J Gynecol Pathol 2020; 39:289-295. [DOI: 10.1097/pgp.0000000000000605] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
16
|
Pompei V, Salvolini E, Rubini C, Lucarini G, Molinelli E, Brisigotti V, Pozzi V, Sartini D, Campanati A, Offidani A, Emanuelli M. Nicotinamide N-methyltransferase in nonmelanoma skin cancers. Eur J Clin Invest 2019; 49:e13175. [PMID: 31571214 DOI: 10.1111/eci.13175] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 09/17/2019] [Accepted: 09/28/2019] [Indexed: 02/02/2023]
Abstract
BACKGROUND Squamous cell carcinoma (SCC) and basal cell carcinoma (BCC) represent the most common forms of nonmelanoma skin cancers (NMSCs). Although successful treatment of these neoplasms is based on surgical excision, an increasing number of BCCs relapses and many SCCs display high rates of recurrence and metastasis. Nicotinamide N-methyltransferase (NNMT) is a cytosolic enzyme, which was found to be upregulated in different solid tumours. However, there are no data regarding enzyme expression in NMSCs. The aim of this study was therefore to evaluate the potential involvement of NNMT in BCCs and SCCs. MATERIALS AND METHODS Immunohistochemical analyses were carried out on 40 BCC cases and 39 SCC cases, to evaluate enzyme expression in tumour and surrounding healthy margins. Moreover, the relationship between NNMT intratumour levels and clinico-pathological parameters were explored. RESULTS Nicotinamide N-methyltransferase was found to be overexpressed in BCCs compared with control tissues, while a significant enzyme downregulation was detected in SCCs with respect to corresponding healthy margins. In addition, NNMT levels were negatively related to aggressiveness of both BCCs (distinguishing between infiltrative and nodular tumours) and SCCs (considering head and neck forms and tumours of the extremities and trunk). CONCLUSIONS These evidences seem to demonstrate that the different NNMT dysregulation detected in BCC and SCC may be the result of important biological traits distinctively characterizing these two forms within NMSCs. In addition, enzyme levels seem to be inversely correlated with tumour aggressiveness, thus suggesting the potential suitability of the enzyme as a prognostic biomarker for both neoplasms.
Collapse
Affiliation(s)
- Veronica Pompei
- Department of Clinical Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Eleonora Salvolini
- Department of Clinical Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Corrado Rubini
- Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, Ancona, Italy
| | - Guendalina Lucarini
- Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Elisa Molinelli
- Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Valerio Brisigotti
- Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Valentina Pozzi
- Department of Clinical Sciences, Polytechnic University of Marche, Ancona, Italy.,New York-Marche Structural Biology Center (NY-MaSBiC), Polytechnic University of Marche, Ancona, Italy
| | - Davide Sartini
- Department of Clinical Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Anna Campanati
- Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Annamaria Offidani
- Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Monica Emanuelli
- Department of Clinical Sciences, Polytechnic University of Marche, Ancona, Italy.,New York-Marche Structural Biology Center (NY-MaSBiC), Polytechnic University of Marche, Ancona, Italy
| |
Collapse
|
17
|
Sartini D, Pompei V, Lucarini G, Rubini C, Molinelli E, Brisigotti V, Salvolini E, Campanati A, Offidani A, Emanuelli M. Differential expression of nicotinamide N-methyltransferase in cutaneous keratoacanthoma and squamous cell carcinoma: an immunohistochemical study. J Eur Acad Dermatol Venereol 2019; 34:e121-e123. [PMID: 31705831 DOI: 10.1111/jdv.16048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- D Sartini
- Department of Clinical Sciences, Marche Polytechnic University, Ancona, Italy
| | - V Pompei
- Department of Clinical Sciences, Marche Polytechnic University, Ancona, Italy
| | - G Lucarini
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - C Rubini
- Department of Biomedical Sciences and Public Health, Marche Polytechnic University, Ancona, Italy
| | - E Molinelli
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - V Brisigotti
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - E Salvolini
- Department of Clinical Sciences, Marche Polytechnic University, Ancona, Italy
| | - A Campanati
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - A Offidani
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - M Emanuelli
- Department of Clinical Sciences, Marche Polytechnic University, Ancona, Italy.,New York-Marche Structural Biology Center (NY-MaSBiC), Marche Polytechnic University, Ancona, Italy
| |
Collapse
|
18
|
Li S, Qiao L, Yang Z, He C. Prognostic Value of Nicotinamide N-Methyltransferase Expression in Patients With Solid Tumors: A Systematic Review and Meta-Analysis. Front Physiol 2018; 9:1407. [PMID: 30349486 PMCID: PMC6187113 DOI: 10.3389/fphys.2018.01407] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 09/14/2018] [Indexed: 12/28/2022] Open
Abstract
Background: Nicotinamide N-methyltransferase (NNMT) is an enzyme that catalyzes N-methylation of pyridine-containing compounds. NNMT is upregulated in many types of solid tumors, suggesting the potential for its use as a tumor biomarker. However, the prognostic value of NNMT in solid tumors is still unclear. We therefore conducted a meta-analysis to investigate the association between NNMT expression and survival in patients with solid tumors. Methods: We focused on patients with solid tumors, using high NNMT expression levels as the intervention and low NNMT expression levels as the comparison, according to Patient, Intervention, Comparison, and Outcome (PICO) guidelines. Electronic databases (up to June 7, 2018) were comprehensively searched to collect relevant cohort studies regarding the associations between NNMT expression levels and survival outcomes (overall survival [OS], disease-specific survival [DSS] including cancer-specific survival [CSS], and time to tumor progression [TTP] including disease-free survival [DFS], progression-free survival [PFS], and metastasis-free survival [MeFS]). Publication biases were also examined. All analyses were performed using STATA 12.0 software. Results: A total of 3340 patients with solid tumors from nine published studies were included. The combined hazard ratio (HR) identified high NNMT expression levels as a poor prognostic predictor of OS (HR = 1.67, 95% CI = 1.23–2.26). However, NNMT levels had no significant association with DSS (HR = 1.47, 95% CI = 0.95–2.28) and TTP (HR = 1.13, 95%CI = 0.39–3.25). Conclusion: High NNMT expression levels may be a poor prognostic biomarker for patients with solid tumors.
Collapse
Affiliation(s)
- Shimeng Li
- Department of Clinical Laboratory, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Lu Qiao
- Department of Clinical Laboratory, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zhaowei Yang
- Department of Clinical Laboratory, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Chengyan He
- Department of Clinical Laboratory, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
19
|
You Z, Liu Y, Liu X. Nicotinamide N-methyltransferase enhances the progression of prostate cancer by stabilizing sirtuin 1. Oncol Lett 2018; 15:9195-9201. [PMID: 29805651 PMCID: PMC5958777 DOI: 10.3892/ol.2018.8474] [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: 08/06/2017] [Accepted: 02/05/2018] [Indexed: 12/12/2022] Open
Abstract
A previous study demonstrated that nicotinamide N-methyltransferase (NNMT) is upregulated in the tissues of patients with prostate cancer (PCa); however, the specific underlying mechanism of this remains unclear. To begin with, the expression of NNMT was investigated in the peripheral blood of patients with PCa and of healthy control subjects. The results indicated that the expression level of NNMT was elevated in the peripheral blood and tissues of patients with PCa. Furthermore, the overexpression of NNMT enhanced PC-3 cell viability, invasion and migration capacity. Additionally, the overexpression of NNMT significantly increased the mRNA level of sirtuin 1 (SIRT1) in PC-3 cells. In addition, nicotinamide treatment significantly suppressed the expression of SIRT1 even in PC-3 cells transfected with adeno-associated virus-NNMT. Furthermore, the PC-3 cell invasion capacity was notably decreased by the nicotinamide treatment; however, such effects were largely abolished by the overexpression of NNMT in PC-3 cells. These data indicated that NNMT enhanced PC-3 cell migration and invasion mainly by regulating SIRT1 expression. In summary, the present study indicated that NNMT is an important regulator of SIRT1 expression in PC-3 cells and may be a potential therapeutic target for PCa.
Collapse
Affiliation(s)
- Zhenyu You
- Department of Oncology, 202 Hospital of Chinese People's Liberation Army, Shenyang, Liaoning 110812, P.R. China
| | - Yang Liu
- Department of Oncology, 202 Hospital of Chinese People's Liberation Army, Shenyang, Liaoning 110812, P.R. China
| | - Xuefei Liu
- Department of Oncology, 202 Hospital of Chinese People's Liberation Army, Shenyang, Liaoning 110812, P.R. China
| |
Collapse
|
20
|
Ganzetti G, Sartini D, Campanati A, Rubini C, Molinelli E, Brisigotti V, Cecati M, Pozzi V, Campagna R, Offidani A, Emanuelli M. Nicotinamide N-methyltransferase: potential involvement in cutaneous malignant melanoma. Melanoma Res 2018; 28:82-88. [PMID: 29420365 DOI: 10.1097/cmr.0000000000000430] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Nicotinamide N-methyltransferase (NNMT) is an enzyme that catalyzes the N-methylation of nicotinamide and pyridine compounds, participating in xenobiotic and drug metabolism. Data on literature have evidenced a possible role of NNMT in many solid cancers, but no data are currently available in cutaneous melanoma. Recent important advances have been achieved in the treatment of advanced melanoma with targeted therapy and immunotherapy. However, the identification of biomarkers that can be used for the detection of early stage disease as well as for monitoring the therapeutic response during treatment is of utmost importance. The aim of this study was to study the possible role of NNMT in melanoma. In the present study, we carried out immunohistochemical analyses to evaluate the expression of the enzyme NNMT in 34 melanomas and 34 nevi. Moreover, we explored the relationship between NNMT levels and the prognostic parameters of patients with melanoma. The results obtained showed significantly (P<0.0001) higher NNMT expression in melanoma compared with that detected in nevi. In addition, a significant (P<0.05) inverse relationship was found between enzyme levels and Breslow thickness, Clark level, the presence/number of mitoses, and ulceration. Taken together, these data seem to suggest that NNMT could represent a molecular biomarker for melanoma, thus highlighting its potential for both diagnosis and prognosis of this neoplasm.
Collapse
Affiliation(s)
| | | | | | - Corrado Rubini
- Biomedical Sciences and Public Health, Polytechnic University of Marche, Ancona, Italy
| | | | | | | | - Valentina Pozzi
- Clinical and Stomatological Sciences
- New York-Marche Structural Biology Centre (NY-MaSBiC), New York City, New York, USA
| | | | | | - Monica Emanuelli
- Clinical and Stomatological Sciences
- New York-Marche Structural Biology Centre (NY-MaSBiC), New York City, New York, USA
| |
Collapse
|
21
|
Conditionally reprogrammed normal and primary tumor prostate epithelial cells: a novel patient-derived cell model for studies of human prostate cancer. Oncotarget 2017; 8:22741-22758. [PMID: 28009986 PMCID: PMC5410259 DOI: 10.18632/oncotarget.13937] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 11/22/2016] [Indexed: 01/16/2023] Open
Abstract
Our previous study demonstrated that conditional reprogramming (CR) allows the establishment of patient-derived normal and tumor epithelial cell cultures from a variety of tissue types including breast, lung, colon and prostate. Using CR, we have established matched normal and tumor cultures, GUMC-29 and GUMC-30 respectively, from a patient's prostatectomy specimen. These CR cells proliferate indefinitely in vitro and retain stable karyotypes. Most importantly, only tumor-derived CR cells (GUMC-30) produced tumors in xenografted SCID mice, demonstrating maintenance of the critical tumor phenotype. Characterization of cells with DNA fingerprinting demonstrated identical patterns in normal and tumor CR cells as well as in xenografted tumors. By flow cytometry, both normal and tumor CR cells expressed basal, luminal, and stem cell markers, with the majority of the normal and tumor CR cells expressing prostate basal cell markers, CD44 and Trop2, as well as luminal marker, CD13, suggesting a transit-amplifying phenotype. Consistent with this phenotype, real time RT-PCR analyses demonstrated that CR cells predominantly expressed high levels of basal cell markers (KRT5, KRT14 and p63), and low levels of luminal markers. When the CR tumor cells were injected into SCID mice, the expression of luminal markers (AR, NKX3.1) increased significantly, while basal cell markers dramatically decreased. These data suggest that CR cells maintain high levels of proliferation and low levels of differentiation in the presence of feeder cells and ROCK inhibitor, but undergo differentiation once injected into SCID mice. Genomic analyses, including SNP and INDEL, identified genes mutated in tumor cells, including components of apoptosis, cell attachment, and hypoxia pathways. The use of matched patient-derived cells provides a unique in vitro model for studies of early prostate cancer.
Collapse
|
22
|
Wang LY, Cui JJ, Zhu T, Shao WH, Zhao Y, Wang S, Zhang YP, Wu JC, Zhang L. Biomarkers identified for prostate cancer patients through genome-scale screening. Oncotarget 2017; 8:92055-92063. [PMID: 29190897 PMCID: PMC5696163 DOI: 10.18632/oncotarget.20739] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 08/07/2017] [Indexed: 12/22/2022] Open
Abstract
Prostate cancer is a threat to men and usually occurs in aged males. Though prostate specific antigen level and Gleason score are utilized for evaluation of the prostate cancer in clinic, the biomarkers for this malignancy have not been widely recognized. Furthermore, the outcome varies across individuals receiving comparable treatment regimens and the underlying mechanism is still unclear. We supposed that genetic feature may be responsible for, at least in part, this process and conducted a two-cohort study to compare the genetic difference in tumorous and normal tissues of prostate cancer patients. The Gene Expression Omnibus dataset were used and a total of 41 genes were found significantly differently expressed in tumor tissues as compared with normal prostate tissues. Four genes (SPOCK3, SPON1, PTN and TGFB3) were selected for further evaluation after Gene Ontology analysis, Kyoto Encyclopedia of Genes and Genomes pathway analysis and clinical association analysis. MIR1908 was also found decreased expression level in prostate cancer whose target genes were found expressing in both prostate tumor and normal tissues. These results indicated that these potential biomarkers deserve attention in prostate cancer patients and the underlying mechanism should be further investigated.
Collapse
Affiliation(s)
- Lei-Yun Wang
- Department of Clinical Pharmacology, XiangYa Hospital, Central South University, Changsha 410008, P.R. China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, P.R. China
| | - Jia-Jia Cui
- Department of Clinical Pharmacology, XiangYa Hospital, Central South University, Changsha 410008, P.R. China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, P.R. China
| | - Tao Zhu
- Department of Clinical Pharmacology, XiangYa Hospital, Central South University, Changsha 410008, P.R. China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, P.R. China
| | - Wei-Hua Shao
- Department of Clinical Pharmacology, XiangYa Hospital, Central South University, Changsha 410008, P.R. China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, P.R. China
| | - Yi Zhao
- Department of Clinical Pharmacology, XiangYa Hospital, Central South University, Changsha 410008, P.R. China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, P.R. China
| | - Sai Wang
- Department of Neurology, XiangYa Hospital, Central South University, Changsha 410008, P.R. China
| | - Yu-Peng Zhang
- Department of Neurology, XiangYa Hospital, Central South University, Changsha 410008, P.R. China
| | - Ji-Chu Wu
- Department of Cardiovascular, Central Hospital of ShaoYang, ShaoYang 422000, P.R. China
| | - Le Zhang
- Department of Neurology, XiangYa Hospital, Central South University, Changsha 410008, P.R. China
| |
Collapse
|
23
|
Ramsden DB, Waring RH, Barlow DJ, Parsons RB. Nicotinamide N-Methyltransferase in Health and Cancer. Int J Tryptophan Res 2017; 10:1178646917691739. [PMID: 35185340 PMCID: PMC8851132 DOI: 10.1177/1178646917691739] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 01/11/2017] [Indexed: 12/19/2022] Open
Abstract
Over the past decade, the roles of nicotinamide N-methyltransferase and its product 1-methyl nicotinamide have emerged from playing merely minor roles in phase 2 xenobiotic metabolism as actors in some of the most important scenes of human life. In this review, the structures of the gene, messenger RNA, and protein are discussed, together with the role of the enzyme in many of the common cancers that afflict people today.
Collapse
Affiliation(s)
- David B Ramsden
- Institute of Metabolism and Systems Research, The Medical School, University of Birmingham, Birmingham, UK
| | | | - David J Barlow
- Institute of Pharmaceutical Science, King’s College London, London, UK
| | - Richard B Parsons
- Institute of Pharmaceutical Science, King’s College London, London, UK
| |
Collapse
|
24
|
Denslow A, Switalska M, Nowak M, Maciejewska M, Chlopicki S, Marcinek A, Gebicki J, Wietrzyk J. The effects of 1,4-dimethylpyridine in metastatic prostate cancer in mice. BMC Cancer 2017; 17:177. [PMID: 28270133 PMCID: PMC5341170 DOI: 10.1186/s12885-017-3161-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 03/01/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND We previously showed that 1-methylnicotinamide (1-MNA) and its analog 1,4-dimethylpyridine (1,4-DMP) could inhibit the formation of lung metastases and enhance the efficacy of cyclophosphamide-based chemotherapy in the model of spontaneously metastasizing 4T1 mouse mammary gland tumors. In the present study, we aimed to investigate whether the previously observed activity of pyridine compounds pertains also to the prevention and the treatment of metastatic prostate tumors, in a combined chemotherapy with docetaxel. METHODS Cancer-preventing activity of 1,4-DMP was studied in the model of prostate tumors spontaneously arising in C57BL/6-Tg (TRAMP)8247Ng/J (TRAMP) mice. The efficacy of the combined chemotherapy, comprising simultaneous use of 1,4-DMP and docetaxel, was evaluated in the orthotopic mouse model of human PC-3M-luc2 prostate cancer. The toxicity of the applied treatment was also determined. RESULTS The development of prostate tumors in TRAMP mice remained unaffected after administration of 1,4-DMP. Similarly, no effect of 1,4-DMP was found on the growth of orthotopically transplanted PC-3M-luc2 tumors. However, when 1,4-DMP was administered along with docetaxel, it enhanced the anticancer activity of the chemotherapy. As a result, in PC-3M-luc2-bearing mice statistically significant inhibition of the tumor growth and lower metastases incidence were observed. The decreased metastatic yield is probably related to the diminished platelet activity observed in mice treated with combined therapeutic regimen. Finally, the combined treatment exhibited lowered side effects accompanying docetaxel administration. CONCLUSIONS Results presented herein confirm previously published data on the anticancer activity of pyridine compounds and demonstrate that 1,4-DMP may be beneficially implemented into chemotherapy utilizing various cytotoxic agents, directed against multiple metastatic tumor types.
Collapse
Affiliation(s)
- Agnieszka Denslow
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, Wroclaw, 53-114 Poland
| | - Marta Switalska
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, Wroclaw, 53-114 Poland
| | - Marcin Nowak
- Wroclaw University of Environmental and Life Sciences, Norwida 31, Wroclaw, 50-375 Poland
| | - Magdalena Maciejewska
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, Wroclaw, 53-114 Poland
| | - Stefan Chlopicki
- Chair of Pharmacology, Jagiellonian University, Medical College, Grzegorzecka 16, Krakow, 31-531 Poland
- Jagiellonian Center for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow, 30-348 Poland
| | - Andrzej Marcinek
- Lodz University of Technology, Zeromskiego 116, Lodz, 90-924 Poland
| | - Jerzy Gebicki
- Lodz University of Technology, Zeromskiego 116, Lodz, 90-924 Poland
| | - Joanna Wietrzyk
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, Wroclaw, 53-114 Poland
| |
Collapse
|
25
|
Aurora Kinase A is a Biomarker for Bladder Cancer Detection and Contributes to its Aggressive Behavior. Sci Rep 2017; 7:40714. [PMID: 28102366 PMCID: PMC5244380 DOI: 10.1038/srep40714] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 12/08/2016] [Indexed: 01/01/2023] Open
Abstract
The effects of AURKA overexpression associated with poor clinical outcomes have been attributed to increased cell cycle progression and the development of genomic instability with aneuploidy. We used RNA interference to examine the effects of AURKA overexpression in human bladder cancer cells. Knockdown had minimal effects on cell proliferation but blocked tumor cell invasion. Whole genome mRNA expression profiling identified nicotinamide N-methyltransferase (NNMT) as a downstream target that was repressed by AURKA. Chromatin immunoprecipitation and NNMT promoter luciferase assays revealed that AURKA’s effects on NNMT were caused by PAX3-mediated transcriptional repression and overexpression of NNMT blocked tumor cell invasion in vitro. Overexpression of AURKA and activation of its downstream pathway was enriched in the basal subtype in primary human tumors and was associated with poor clinical outcomes. We also show that the FISH test for the AURKA gene copy number in urine yielded a specificity of 79.7% (95% confidence interval [CI] = 74.2% to 84.1%), and a sensitivity of 79.6% (95% CI = 74.2% to 84.1%) with an AUC of 0.901 (95% CI = 0.872 to 0.928; P < 0.001). These results implicate AURKA as an effective biomarker for bladder cancer detection as well as therapeutic target especially for its basal type.
Collapse
|
26
|
Palanichamy K, Kanji S, Gordon N, Thirumoorthy K, Jacob JR, Litzenberg KT, Patel D, Chakravarti A. NNMT Silencing Activates Tumor Suppressor PP2A, Inactivates Oncogenic STKs, and Inhibits Tumor Forming Ability. Clin Cancer Res 2016; 23:2325-2334. [PMID: 27810903 DOI: 10.1158/1078-0432.ccr-16-1323] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 09/19/2016] [Accepted: 10/12/2016] [Indexed: 12/12/2022]
Abstract
Purpose: To identify potential molecular hubs that regulate oncogenic kinases and target them to improve treatment outcomes for glioblastoma patients.Experimental Design: Data mining of The Cancer Genome Atlas datasets identified nicotinamide-N-methyl transferase (NNMT) as a prognostic marker for glioblastoma, an enzyme linked to the reorganization of the methylome. We tested our hypothesis that NNMT plays a crucial role by modulating protein methylation, leading to inactivation of tumor suppressors and activation of oncogenes. Further experiments were performed to understand the underlying biochemical mechanisms using glioblastoma patient samples, established, primary, and isogenic cells.Results: We demonstrate that NNMT outcompetes leucine carboxyl methyl transferase 1 (LCMT1) for methyl transfer from principal methyl donor SAM in biological systems. Inhibiting NNMT increased the availability of methyl groups for LCMT1 to methylate PP2A, resulting in the inhibition of oncogenic serine/threonine kinases (STK). Further, NNMT inhibition retained the radiosensitizer nicotinamide and enhanced radiation sensitivity. We have provided the biochemical rationale of how NNMT plays a vital role in inhibiting tumor suppressor PP2A while concomitantly activating STKs.Conclusions: We report the intricate novel mechanism in which NNMT inhibits tumor suppressor PP2A by reorganizing the methylome both at epigenome and proteome levels and concomitantly activating prosurvival STKs. In glioblastoma tumors with NNMT expression, activation of PP2A can be accomplished by FDA approved perphenazine (PPZ), which is currently used to treat mood disorders such as schizophrenia, bipolar disorder, etc. This study forms a foundation for further glioblastoma clinical trials using PPZ with standard of care treatment. Clin Cancer Res; 23(9); 2325-34. ©2016 AACR.
Collapse
Affiliation(s)
- Kamalakannan Palanichamy
- Department of Radiation Oncology, James Cancer Hospital and Solove Research Institute, The Ohio State University College of Medicine and Comprehensive Cancer Center, Columbus, Ohio.
| | - Suman Kanji
- Department of Radiation Oncology, James Cancer Hospital and Solove Research Institute, The Ohio State University College of Medicine and Comprehensive Cancer Center, Columbus, Ohio
| | - Nicolaus Gordon
- Department of Radiation Oncology, James Cancer Hospital and Solove Research Institute, The Ohio State University College of Medicine and Comprehensive Cancer Center, Columbus, Ohio
| | - Krishnan Thirumoorthy
- Department of Radiation Oncology, James Cancer Hospital and Solove Research Institute, The Ohio State University College of Medicine and Comprehensive Cancer Center, Columbus, Ohio
| | - John R Jacob
- Department of Radiation Oncology, James Cancer Hospital and Solove Research Institute, The Ohio State University College of Medicine and Comprehensive Cancer Center, Columbus, Ohio
| | - Kevin T Litzenberg
- Department of Radiation Oncology, James Cancer Hospital and Solove Research Institute, The Ohio State University College of Medicine and Comprehensive Cancer Center, Columbus, Ohio
| | - Disha Patel
- Department of Radiation Oncology, James Cancer Hospital and Solove Research Institute, The Ohio State University College of Medicine and Comprehensive Cancer Center, Columbus, Ohio
| | - Arnab Chakravarti
- Department of Radiation Oncology, James Cancer Hospital and Solove Research Institute, The Ohio State University College of Medicine and Comprehensive Cancer Center, Columbus, Ohio
| |
Collapse
|
27
|
Drew JE, Farquharson AJ, Horgan GW, Williams LM. Tissue-specific regulation of sirtuin and nicotinamide adenine dinucleotide biosynthetic pathways identified in C57Bl/6 mice in response to high-fat feeding. J Nutr Biochem 2016; 37:20-29. [PMID: 27592202 DOI: 10.1016/j.jnutbio.2016.07.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 06/16/2016] [Accepted: 07/15/2016] [Indexed: 01/21/2023]
Abstract
The sirtuin (SIRT)/nicotinamide adenine dinucleotide (NAD) system is implicated in development of type 2 diabetes (T2D) and diet-induced obesity, a major risk factor for T2D. Mechanistic links have not yet been defined. SIRT/NAD system gene expression and NAD/NADH levels were measured in liver, white adipose tissue (WAT) and skeletal muscle from mice fed either a low-fat diet or high-fat diet (HFD) for 3 days up to 16 weeks. An in-house custom-designed multiplex gene expression assay assessed all 7 mouse SIRTs (SIRT1-7) and 16 enzymes involved in conversion of tryptophan, niacin, nicotinamide riboside and metabolic precursors to NAD. Significantly altered transcription was correlated with body weight, fat mass, plasma lipids and hormones. Regulation of the SIRT/NAD system was associated with early (SIRT4, SIRT7, NAPRT1 and NMNAT2) and late phases (NMNAT3, NMRK2, ABCA1 and CD38) of glucose intolerance. TDO2 and NNMT were identified as markers of HFD consumption. Altered regulation of the SIRT/NAD system in response to HFD was prominent in liver compared with WAT or muscle. Multiple components of the SIRTs and NAD biosynthetic enzymes network respond to consumption of dietary fat. Novel molecular targets identified above could direct strategies for dietary/therapeutic interventions to limit metabolic dysfunction and development of T2D.
Collapse
Affiliation(s)
- Janice E Drew
- Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen AB21 9SB, Scotland.
| | - Andrew J Farquharson
- Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen AB21 9SB, Scotland
| | - Graham W Horgan
- Biomathematics and Statistics Scotland, Aberdeen AB21 9SB, Scotland
| | - Lynda M Williams
- Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen AB21 9SB, Scotland
| |
Collapse
|
28
|
Blazejczyk A, Switalska M, Chlopicki S, Marcinek A, Gebicki J, Nowak M, Nasulewicz-Goldeman A, Wietrzyk J. 1-methylnicotinamide and its structural analog 1,4-dimethylpyridine for the prevention of cancer metastasis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2016; 35:110. [PMID: 27412454 PMCID: PMC4944260 DOI: 10.1186/s13046-016-0389-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 06/28/2016] [Indexed: 12/16/2022]
Abstract
Background 1-methylnicotinamide (1-MNA), an endogenous metabolite of nicotinamide, has recently gained interest due to its anti-inflammatory and anti-thrombotic activities linked to the COX-2/PGI2 pathway. Given the previously reported anti-metastatic activity of prostacyclin (PGI2), we aimed to assess the effects of 1-MNA and its structurally related analog, 1,4-dimethylpyridine (1,4-DMP), in the prevention of cancer metastasis. Methods All the studies on the anti-tumor and anti-metastatic activity of 1-MNA and 1,4-DMP were conducted using the model of murine mammary gland cancer (4T1) transplanted either orthotopically or intravenously into female BALB/c mouse. Additionally, the effect of the investigated molecules on cancer cell-induced angiogenesis was estimated using the matrigel plug assay utilizing 4T1 cells as a source of pro-angiogenic factors. Results Neither 1-MNA nor 1,4-DMP, when given in a monotherapy of metastatic cancer, influenced the growth of 4T1 primary tumors transplanted orthotopically; however, both compounds tended to inhibit 4T1 metastases formation in lungs of mice that were orthotopically or intravenously inoculated with 4T1 or 4T1-luc2-tdTomato cells, respectively. Additionally, while 1-MNA enhanced tumor vasculature formation and markedly increased PGI2 generation, 1,4-DMP did not have such an effect. The anti-metastatic activity of 1-MNA and 1,4-DMP was further confirmed when both agents were applied with a cytostatic drug in a combined treatment of 4T1 murine mammary gland cancer what resulted in up to 80 % diminution of lung metastases formation. Conclusions The results of the studies presented below indicate that 1-MNA and its structural analog 1,4-DMP prevent metastasis and might be beneficially implemented into the treatment of metastatic breast cancer to ensure a comprehensive strategy of metastasis control. Electronic supplementary material The online version of this article (doi:10.1186/s13046-016-0389-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Agnieszka Blazejczyk
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114, Wroclaw, Poland
| | - Marta Switalska
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114, Wroclaw, Poland
| | - Stefan Chlopicki
- Chair of Pharmacology, Jagiellonian University, Medical College, Grzegórzecka 16, 31-531, Krakow, Poland.,Jagiellonian Center for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348, Krakow, Poland
| | - Andrzej Marcinek
- Lodz University of Technology, Zeromskiego 116, 90-924, Lodz, Poland
| | - Jerzy Gebicki
- Lodz University of Technology, Zeromskiego 116, 90-924, Lodz, Poland
| | - Marcin Nowak
- Wroclaw University of Environmental and Life Sciences, Norwida 31, 50-375, Wroclaw, Poland
| | - Anna Nasulewicz-Goldeman
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114, Wroclaw, Poland
| | - Joanna Wietrzyk
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114, Wroclaw, Poland.
| |
Collapse
|