1
|
Sun WD, Zhu XJ, Li JJ, Mei YZ, Li WS, Li JH. Nicotinamide N-methyltransferase (NNMT): A key enzyme in cancer metabolism and therapeutic target. Int Immunopharmacol 2024; 142:113208. [PMID: 39312861 DOI: 10.1016/j.intimp.2024.113208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/17/2024] [Accepted: 09/17/2024] [Indexed: 09/25/2024]
Abstract
Emerging research has positioned Nicotinamide N-methyltransferase (NNMT) as a key player in oncology, with its heightened expression frequently observed across diverse cancers. This increased presence is tightly linked to tumor initiation, proliferation, and metastasis. The enzymatic function of NNMT is centered on the methylation of nicotinamide (NAM), utilizing S-adenosylmethionine (SAM) as the methyl donor, which results in the generation of S-adenosyl-L-homocysteine (SAH) and methyl nicotinamide (MNAM). This metabolic process reduces the availability of NAM, necessary for Nicotinamide adenine dinucleotide (NAD+) synthesis, and generates SAH, precursor to homocysteine (Hcy). These alterations are theorized to foster the resilience, expansion, and invasiveness of cancer cells. Furthermore, NNMT is implicated in enhancing cancer malignancy by affecting multiple signaling pathways, such as phosphatidylinositol 3-kinase (PI3K)-protein kinase B (AKT), cancer-associated fibroblasts (CAFs) and 5-Methyladenosine (5-MA), epithelial-mesenchymal transition (EMT), and epigenetic mechanisms. Upregulation of NNMT metabolism plays a key role in the formation and maintenance of the tumour microenvironment. While the use of small molecule inhibitors and RNA interference (RNAi) to target NNMT has shown therapeutic promise, the full extent of NNMT's influence on cancer is not yet fully understood, and clinical evidence is limited. This article systematically describes the relationship between the functional metabolism of NNMT enzymes and the cancer and tumour microenvironments, describing the mechanisms by which NNMT contributes to cancer initiation, proliferation, and metastasis, as well as targeted therapies. Additionally, we discuss the future opportunities and challenges of NNMT in targeted anti-cancer treatments.
Collapse
Affiliation(s)
- Wei-Dong Sun
- Key Lab of Aquatic Training Monitoring and Intervention of General Administration of Sport of China, Physical Education College, Jiangxi Normal University, Nanchang 330022, Jiangxi Province, China
| | - Xiao-Juan Zhu
- Key Lab of Aquatic Training Monitoring and Intervention of General Administration of Sport of China, Physical Education College, Jiangxi Normal University, Nanchang 330022, Jiangxi Province, China
| | - Jing-Jing Li
- Key Lab of Aquatic Training Monitoring and Intervention of General Administration of Sport of China, Physical Education College, Jiangxi Normal University, Nanchang 330022, Jiangxi Province, China
| | - Ya-Zhong Mei
- Key Lab of Aquatic Training Monitoring and Intervention of General Administration of Sport of China, Physical Education College, Jiangxi Normal University, Nanchang 330022, Jiangxi Province, China
| | - Wen-Song Li
- Key Lab of Aquatic Training Monitoring and Intervention of General Administration of Sport of China, Physical Education College, Jiangxi Normal University, Nanchang 330022, Jiangxi Province, China
| | - Jiang-Hua Li
- Key Lab of Aquatic Training Monitoring and Intervention of General Administration of Sport of China, Physical Education College, Jiangxi Normal University, Nanchang 330022, Jiangxi Province, China.
| |
Collapse
|
2
|
Liu YN, Liu MK, Wen YC, Li CH, Yeh HL, Dung PVT, Jiang KC, Chen WH, Li HR, Huang J, Chen WY. Binding of interleukin-1 receptor antagonist to cholinergic receptor muscarinic 4 promotes immunosuppression and neuroendocrine differentiation in prostate cancer. Cancer Lett 2024; 598:217090. [PMID: 38945201 DOI: 10.1016/j.canlet.2024.217090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/14/2024] [Accepted: 06/25/2024] [Indexed: 07/02/2024]
Abstract
The tumor microenvironment (TME) of prostate cancer (PCa) is characterized by high levels of immunosuppressive molecules, including cytokines and chemokines. This creates a hostile immune landscape that impedes effective immune responses. The interleukin-1 (IL-1) receptor antagonist (IL1RN), a key anti-inflammatory molecule, plays a significant role in suppressing IL-1-related immune and inflammatory responses. Our research investigates the oncogenic role of IL1RN in PCa, particularly its interactions with muscarinic acetylcholine receptor 4 (CHRM4), and its involvement in driving immunosuppressive pathways and M2-like macrophage polarization within the PCa TME. We demonstrate that following androgen deprivation therapy (ADT), the IL1RN-CHRM4 interaction in PCa activates the MAPK/AKT signaling pathway. This activation upregulates the transcription factors E2F1 and MYCN, stimulating IL1RN production and creating a positive feedback loop that increases CHRM4 abundance in both PCa cells and M2-like macrophages. This ADT-driven IL1RN/CHRM4 axis significantly enhances immune checkpoint markers associated with neuroendocrine differentiation and treatment-resistant outcomes. Higher serum IL1RN levels are associated with increased disease aggressiveness and M2-like macrophage markers in advanced PCa patients. Additionally, elevated IL1RN levels correlate with better clinical outcomes following immunotherapy. Clinical correlations between IL1RN and CHRM4 expression in advanced PCa patients and neuroendocrine PCa organoid models highlight their potential as therapeutic targets. Our data suggest that targeting the IL1RN/CHRM4 signaling could be a promising strategy for managing PCa progression and enhancing treatment responses.
Collapse
Affiliation(s)
- Yen-Nien Liu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Ming-Kun Liu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Yu-Ching Wen
- Department of Urology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Urology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei, Taiwan
| | - Chien-Hsiu Li
- Department of Urology, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan
| | - Hsiu-Lien Yeh
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Phan Vu Thuy Dung
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Kuo-Ching Jiang
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Wei-Hao Chen
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Han-Ru Li
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Jiaoti Huang
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Wei-Yu Chen
- Department of Pathology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
3
|
Cheng YC, Acedera JD, Li YJ, Shieh SY. A keratinocyte-adipocyte signaling loop is reprogrammed by loss of BTG3 to augment skin carcinogenesis. Cell Death Differ 2024; 31:970-982. [PMID: 38714880 PMCID: PMC11303697 DOI: 10.1038/s41418-024-01304-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 08/09/2024] Open
Abstract
Obesity is endemic to many developed countries. Overweight or obesity is associated with an increased risk of developing cancer. Dysfunctional adipose tissue alters cancer cell proliferation and migration; however, whether and how neoplastic epithelial cells communicate with adipose tissue and the underlying mechanism are less clear. BTG3 is a member of the anti-proliferative BTG/Tob family and functions as a tumor suppressor. Here, we demonstrated that BTG3 levels are downregulated in basal cell carcinoma and squamous cell carcinoma compared to normal skin tissue, and Btg3 knockout in mice augmented the development of papilloma in a mouse model of DMBA/TPA-induced skin carcinogenesis. Mechanistically, BTG3-knockout keratinocytes promoted adipocyte differentiation mainly through the release of IL1α, IL10, and CCL4, as a result of elevated NF-κB activity. These adipocytes produced CCL20 and FGF7 in a feedback loop to promote keratinocyte migration. Thus, our findings showcased the role of BTG3 in guarding the interplay between keratinocytes and adjacent adipocytes, and identified the underlying neoplastic molecular mediators that may serve as possible targets in the treatment of skin cancer.
Collapse
Affiliation(s)
- Yu-Che Cheng
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Jack Dalit Acedera
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| | - Yi-Ju Li
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Sheau-Yann Shieh
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
4
|
Li JJ, Sun WD, Zhu XJ, Mei YZ, Li WS, Li JH. Nicotinamide N-Methyltransferase (NNMT): A New Hope for Treating Aging and Age-Related Conditions. Metabolites 2024; 14:343. [PMID: 38921477 PMCID: PMC11205546 DOI: 10.3390/metabo14060343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/09/2024] [Accepted: 06/10/2024] [Indexed: 06/27/2024] Open
Abstract
The complex process of aging leads to a gradual deterioration in the function of cells, tissues, and the entire organism, thereby increasing the risk of disease and death. Nicotinamide N-methyltransferase (NNMT) has attracted attention as a potential target for combating aging and its related pathologies. Studies have shown that NNMT activity increases over time, which is closely associated with the onset and progression of age-related diseases. NNMT uses S-adenosylmethionine (SAM) as a methyl donor to facilitate the methylation of nicotinamide (NAM), converting NAM into S-adenosyl-L-homocysteine (SAH) and methylnicotinamide (MNA). This enzymatic action depletes NAM, a precursor of nicotinamide adenine dinucleotide (NAD+), and generates SAH, a precursor of homocysteine (Hcy). The reduction in the NAD+ levels and the increase in the Hcy levels are considered important factors in the aging process and age-related diseases. The efficacy of RNA interference (RNAi) therapies and small-molecule inhibitors targeting NNMT demonstrates the potential of NNMT as a therapeutic target. Despite these advances, the exact mechanisms by which NNMT influences aging and age-related diseases remain unclear, and there is a lack of clinical trials involving NNMT inhibitors and RNAi drugs. Therefore, more in-depth research is needed to elucidate the precise functions of NNMT in aging and promote the development of targeted pharmaceutical interventions. This paper aims to explore the specific role of NNMT in aging, and to evaluate its potential as a therapeutic target.
Collapse
Affiliation(s)
| | | | | | | | | | - Jiang-Hua Li
- Physical Education College, Jiangxi Normal University, Nanchang 330022, China; (J.-J.L.); (W.-D.S.); (X.-J.Z.); (Y.-Z.M.); (W.-S.L.)
| |
Collapse
|
5
|
Liu YN, Chen WY, Yeh HL, Chen WH, Jiang KC, Li HR, Dung PVT, Chen ZQ, Lee WJ, Hsiao M, Huang J, Wen YC. MCTP1 increases the malignancy of androgen-deprived prostate cancer cells by inducing neuroendocrine differentiation and EMT. Sci Signal 2024; 17:eadc9142. [PMID: 38861615 DOI: 10.1126/scisignal.adc9142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 05/23/2024] [Indexed: 06/13/2024]
Abstract
Neuroendocrine prostate cancer (PCa) (NEPC), an aggressive subtype that is associated with poor prognosis, may arise after androgen deprivation therapy (ADT). We investigated the molecular mechanisms by which ADT induces neuroendocrine differentiation in advanced PCa. We found that transmembrane protein 1 (MCTP1), which has putative Ca2+ sensing function and multiple Ca2+-binding C2 domains, was abundant in samples from patients with advanced PCa. MCTP1 was associated with the expression of the EMT-associated transcription factors ZBTB46, FOXA2, and HIF1A. The increased abundance of MCTP1 promoted PC3 prostate cancer cell migration and neuroendocrine differentiation and was associated with SNAI1-dependent EMT in C4-2 PCa cells after ADT. ZBTB46 interacted with FOXA2 and HIF1A and increased the abundance of MCTP1 in a hypoxia-dependent manner. MCTP1 stimulated Ca2+ signaling and AKT activation to promote EMT and neuroendocrine differentiation by increasing the SNAI1-dependent expression of EMT and neuroendocrine markers, effects that were blocked by knockdown of MCTP1. These data suggest an oncogenic role for MCTP1 in the maintenance of a rare and aggressive prostate cancer subtype through its response to Ca2+ and suggest its potential as a therapeutic target.
Collapse
Affiliation(s)
- Yen-Nien Liu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Wei-Yu Chen
- Department of Pathology, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Hsiu-Lien Yeh
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Wei-Hao Chen
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Kuo-Ching Jiang
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Han-Ru Li
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Phan Vu Thuy Dung
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Zi-Qing Chen
- Division of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan
| | - Wei-Jiunn Lee
- Department of Urology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
- Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Jiaoti Huang
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Yu-Ching Wen
- Department of Urology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Department of Urology, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
6
|
Serritelli EN, Sartini D, Campagna R, Pozzi V, Martin NI, van Haren MJ, Salvolini E, Cecati M, Rubini C, Emanuelli M. Targeting nicotinamide N-methyltransferase decreased aggressiveness of osteosarcoma cells. Eur J Clin Invest 2024; 54:e14185. [PMID: 38426563 DOI: 10.1111/eci.14185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 02/07/2024] [Accepted: 02/12/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND Osteosarcoma (OS) is a primary bone malignancy that mostly affects young people, characterized by high metastatic potential, and a marked chemoresistance that is responsible for disease relapse in most patients. Therefore, it is necessary to identify novel molecules to setup targeted strategies to improve the clinical outcome. The enzyme nicotinamide N-methyltransferase (NNMT) catalyses the N-methylation of nicotinamide and other analogs, playing a crucial role in the biotransformation of drugs and xenobiotics. NNMT overexpression was reported in a wide variety of cancers, and several studies demonstrated that is able to promote cell proliferation, migration and resistance to chemotherapy. The aim of this study was to explore the potential involvement of NNMT in OS. METHODS Immunohistochemical analyses have been performed to evaluate NNMT expression in selected OS and healthy bone tissue samples. Subsequently, OS cell lines have been transfected with vectors targeting NNMT mRNA (shRNAs) and the impact of this downregulation on migration, cell proliferation, and response to chemotherapeutic treatment was also analysed by wound healing, MTT, SRB and Trypan blue assays, respectively. RESULTS Results showed that OS samples display a significantly higher NNMT expression compared with healthy tissue. Preliminary results suggest that NNMT silencing in OS cell lines is associated to a decrease of cell proliferation and migration, as well as to enhanced sensitivity to chemotherapy. Data obtained showed that NNMT may represent an interesting marker for OS detection and a promising target for effective anti-cancer therapy.
Collapse
Affiliation(s)
- E N Serritelli
- Department of Clinical Sciences, Polytechnic University of Marche, Ancona, Italy
| | - D Sartini
- Department of Clinical Sciences, Polytechnic University of Marche, Ancona, Italy
| | - R Campagna
- Department of Clinical Sciences, Polytechnic University of Marche, Ancona, Italy
| | - V Pozzi
- Department of Clinical Sciences, Polytechnic University of Marche, Ancona, Italy
| | - N I Martin
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, Leiden, the Netherlands
| | - M J van Haren
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, Leiden, the Netherlands
| | - E Salvolini
- Department of Clinical Sciences, Polytechnic University of Marche, Ancona, Italy
| | - M Cecati
- Department of Clinical Sciences, Polytechnic University of Marche, Ancona, Italy
| | - C Rubini
- Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, Ancona, Italy
| | - M 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
|
7
|
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] [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
|
8
|
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: 3.0] [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
|
9
|
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: 7] [Impact Index Per Article: 7.0] [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
|
10
|
Iyamu ID, Zhao T, Huang R. Structure-Activity Relationship Studies on Cell-Potent Nicotinamide N-Methyltransferase Bisubstrate Inhibitors. J Med Chem 2023; 66:10510-10527. [PMID: 37523719 DOI: 10.1021/acs.jmedchem.3c00632] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
Nicotinamide N-methyltransferase (NNMT) is a metabolic enzyme implicated in multiple diseases, making it a promising therapeutic target. Building upon our recently reported NNMT inhibitor II399, we systematically investigate the structure-activity relationship by designing and synthesizing a series of analogues. Among them, two top inhibitors II559 (Ki = 1.2 nM) and II802 (Ki = 1.6 nM) displayed over 5000-fold selectivity for NNMT over closely related methyltransferases. Moreover, II559 and II802 showed enhanced cellular inhibition, with a cellular IC50 value of approximately 150 nM, making them the most cell-potent bisubstrate inhibitors reported to date. Furthermore, both inhibitors reduced the cell viability with a GI50 value of ∼10 μM and suppressed the migration of aggressive clear cell renal cancer cell carcinoma cell lines. Overall, II559 and II802 would serve as valuable probes to investigate the enzymatic function of NNMT in health and diseases.
Collapse
Affiliation(s)
- Iredia D Iyamu
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Institute for Drug Discovery, Purdue University Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907, United States
| | - Tianqi Zhao
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Institute for Drug Discovery, Purdue University Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907, United States
| | - Rong Huang
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Institute for Drug Discovery, Purdue University Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
11
|
Couto JP, Vulin M, Jehanno C, Coissieux M, Hamelin B, Schmidt A, Ivanek R, Sethi A, Bräutigam K, Frei AL, Hager C, Manivannan M, Gómez‐Miragaya J, Obradović MMS, Varga Z, Koelzer VH, Mertz KD, Bentires‐Alj M. Nicotinamide N-methyltransferase sustains a core epigenetic program that promotes metastatic colonization in breast cancer. EMBO J 2023; 42:e112559. [PMID: 37259596 PMCID: PMC10308372 DOI: 10.15252/embj.2022112559] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 04/25/2023] [Accepted: 05/05/2023] [Indexed: 06/02/2023] Open
Abstract
Metastatic colonization of distant organs accounts for over 90% of deaths related to solid cancers, yet the molecular determinants of metastasis remain poorly understood. Here, we unveil a mechanism of colonization in the aggressive basal-like subtype of breast cancer that is driven by the NAD+ metabolic enzyme nicotinamide N-methyltransferase (NNMT). We demonstrate that NNMT imprints a basal genetic program into cancer cells, enhancing their plasticity. In line, NNMT expression is associated with poor clinical outcomes in patients with breast cancer. Accordingly, ablation of NNMT dramatically suppresses metastasis formation in pre-clinical mouse models. Mechanistically, NNMT depletion results in a methyl overflow that increases histone H3K9 trimethylation (H3K9me3) and DNA methylation at the promoters of PR/SET Domain-5 (PRDM5) and extracellular matrix-related genes. PRDM5 emerged in this study as a pro-metastatic gene acting via induction of cancer-cell intrinsic transcription of collagens. Depletion of PRDM5 in tumor cells decreases COL1A1 deposition and impairs metastatic colonization of the lungs. These findings reveal a critical activity of the NNMT-PRDM5-COL1A1 axis for cancer cell plasticity and metastasis in basal-like breast cancer.
Collapse
Affiliation(s)
- Joana Pinto Couto
- Department of Biomedicine, University Hospital BaselUniversity of BaselBaselSwitzerland
- Department of SurgeryUniversity Hospital BaselBaselSwitzerland
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
| | - Milica Vulin
- Department of Biomedicine, University Hospital BaselUniversity of BaselBaselSwitzerland
- Department of SurgeryUniversity Hospital BaselBaselSwitzerland
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
| | - Charly Jehanno
- Department of Biomedicine, University Hospital BaselUniversity of BaselBaselSwitzerland
- Department of SurgeryUniversity Hospital BaselBaselSwitzerland
| | - Marie‐May Coissieux
- Department of Biomedicine, University Hospital BaselUniversity of BaselBaselSwitzerland
- Department of SurgeryUniversity Hospital BaselBaselSwitzerland
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
| | - Baptiste Hamelin
- Department of Biomedicine, University Hospital BaselUniversity of BaselBaselSwitzerland
- Department of SurgeryUniversity Hospital BaselBaselSwitzerland
| | - Alexander Schmidt
- Proteomics Core Facility, BiozentrumUniversity of BaselBaselSwitzerland
| | - Robert Ivanek
- Department of Biomedicine, University Hospital BaselUniversity of BaselBaselSwitzerland
- Swiss Institute of BioinformaticsBaselSwitzerland
| | - Atul Sethi
- Department of Biomedicine, University Hospital BaselUniversity of BaselBaselSwitzerland
- Department of SurgeryUniversity Hospital BaselBaselSwitzerland
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
- Swiss Institute of BioinformaticsBaselSwitzerland
| | - Konstantin Bräutigam
- Computational and Translational Pathology Group, Department of Pathology and Molecular Pathology, University Hospital ZurichUniversity of ZurichZürichSwitzerland
- Institute of PathologyUniversity of BernBernSwitzerland
| | - Anja L Frei
- Computational and Translational Pathology Group, Department of Pathology and Molecular Pathology, University Hospital ZurichUniversity of ZurichZürichSwitzerland
| | - Carolina Hager
- Department of Biomedicine, University Hospital BaselUniversity of BaselBaselSwitzerland
- Department of SurgeryUniversity Hospital BaselBaselSwitzerland
| | - Madhuri Manivannan
- Department of Biomedicine, University Hospital BaselUniversity of BaselBaselSwitzerland
- Department of SurgeryUniversity Hospital BaselBaselSwitzerland
| | - Jorge Gómez‐Miragaya
- Department of Biomedicine, University Hospital BaselUniversity of BaselBaselSwitzerland
- Department of SurgeryUniversity Hospital BaselBaselSwitzerland
| | - Milan MS Obradović
- Department of Biomedicine, University Hospital BaselUniversity of BaselBaselSwitzerland
- Department of SurgeryUniversity Hospital BaselBaselSwitzerland
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
| | - Zsuzsanna Varga
- Computational and Translational Pathology Group, Department of Pathology and Molecular Pathology, University Hospital ZurichUniversity of ZurichZürichSwitzerland
| | - Viktor H Koelzer
- Computational and Translational Pathology Group, Department of Pathology and Molecular Pathology, University Hospital ZurichUniversity of ZurichZürichSwitzerland
| | - Kirsten D Mertz
- Institute of PathologyCantonal Hospital BasellandLiestalSwitzerland
| | - Mohamed Bentires‐Alj
- Department of Biomedicine, University Hospital BaselUniversity of BaselBaselSwitzerland
- Department of SurgeryUniversity Hospital BaselBaselSwitzerland
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
| |
Collapse
|
12
|
TRIM56 acts through the IQGAP1-CDC42 signaling axis to promote glioma cell migration and invasion. Cell Death Dis 2023; 14:178. [PMID: 36870986 PMCID: PMC9985612 DOI: 10.1038/s41419-023-05702-6] [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: 10/20/2022] [Revised: 02/19/2023] [Accepted: 02/21/2023] [Indexed: 03/06/2023]
Abstract
Diffuse invasion is an important factor leading to treatment resistance and a poor prognosis in gliomas. Herein, we found that expression of the tripartite motif containing 56 (TRIM56), a RING-finger domain containing E3 ubiquitin ligase, was markedly higher in glioma than in normal brain tissue, and was significantly correlated with malignant phenotypes and a poor prognosis. In vitro and in vivo experimental studies revealed that TRIM56 promoted the migration and invasion of glioma cells. Mechanistically, TRIM56 was transcriptionally regulated by SP1 and promoted the K48-K63-linked poly-ubiquitination transition of IQGAP1 at Lys-1230 by interacting with it, which in turn promoted CDC42 activation. This mechanism was confirmed to mediate glioma migration and invasion. In conclusion, our study provides insights into the mechanisms through which TRIM56 promotes glioma motility, i.e., by regulating IQGAP1 ubiquitination to promote CDC42 activation, which might be clinically targeted for the treatment of glioma.
Collapse
|
13
|
Koch KC, Tew GN. Functional antibody delivery: Advances in cellular manipulation. Adv Drug Deliv Rev 2023; 192:114586. [PMID: 36280179 DOI: 10.1016/j.addr.2022.114586] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/10/2022] [Accepted: 10/18/2022] [Indexed: 02/03/2023]
Abstract
The current therapeutic antibody market in the U.S. consists of 100 antibody-based products and their market value is expected to explode beyond $300 billion by 2025. These therapies are presently limited to extracellular targets due to the innate inability of antibodies to transverse membranes. To expand the number of accessible therapeutic targets, intracellular antibody delivery is necessary. Many delivery vehicles for antibodies have been used with some promising results, such as nanoparticles and cell penetrating polymers. Despite the success of these delivery platforms using model antibody cargo, there is a surprisingly small number of studies that focus on functional antibody delivery into the cytosol that also measures a cellular response. Antibodies can be designed for essentially unlimited targets, including proteins and DNA, that will ultimately control cell function once delivered inside cells. Advancement in cellular manipulation depends on the application of intracellularly delivering functional antibodies to achieve a desired result. This review focuses on the emerging field of functional antibody delivery which enables various cellular responses and cell manipulation.
Collapse
Affiliation(s)
- Kayla C Koch
- Department of Polymer Science & Engineering, University of Massachusetts, Amherst, MA 01003, United States
| | - Gregory N Tew
- Department of Polymer Science & Engineering, University of Massachusetts, Amherst, MA 01003, United States; Molecular & Cellular Biology Program, University of Massachusetts, Amherst, MA 01003, United States; Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, MA 01003, United States.
| |
Collapse
|
14
|
Erden Tayhan S, Bilgin S, Yıldırım A, Koç E. Biological Screening of Polyphenol Derivatives for Anti-Proliferative, Anti-Apoptotic and Anti-Migrative Activities in Human Breast Cancer Cell Lines MCF-7. Chem Biodivers 2023; 20:e202200872. [PMID: 36594615 DOI: 10.1002/cbdv.202200872] [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: 09/16/2022] [Accepted: 12/05/2022] [Indexed: 01/04/2023]
Abstract
Breast cancer is known as the most common type of invasive cancer in women. It is well-known that phenolic compounds play an important role in the treatment of this disease. This study hypothesized that isoeugenol based two polyphenolic compounds 1 and 2 exerts its anti-proliferative effects through the induction of apoptosis and cell migration arrest on human breast cancer cell. Based on this hypothesis, the study aimed to investigate the anti-proliferative, anti-migrative effects of these compounds and their possible basic molecular mechanisms of action in MCF-7 cell lines. As a result, isoeugenol-based compounds 1 and 2 showed anti-proliferative, anti-apoptotic and anti-migrative effects in MCF-7 breast cancer cells. This result was supported by molecular analyzes and it was determined that there were changes in the expression of some gene regions involved in apoptosis and migration. Additionally, it was a remarkable result that cell viability inhibition did not occur in healthy breast tissue cells and no cytotoxic effect was observed. The existence of such a differentiation between cancer cells and healthy cells significantly increases the potential of these compounds to be used as chemotherapeutic drug active ingredients without side effects.
Collapse
Affiliation(s)
- Seçil Erden Tayhan
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tokat Gaziosmanpasa University, 60250, Tokat, Turkey
| | - Sema Bilgin
- Department of Medical Laboratory Techniques, Vocational School of Health Services, Tokat Gaziosmanpasa University, 60250, Tokat, Turkey
| | - Aslı Yıldırım
- Department of Bioengineering, Institute of Graduate Studies, Tokat Gaziosmanpasa University, 60250, Tokat, Turkey
| | - Esra Koç
- Department of Chemistry, Faculty of Arts and Sciences, Tokat Gaziosmanpasa University, 60250, Tokat, Turkey
| |
Collapse
|
15
|
Schwager SC, Young KM, Hapach LA, Carlson CM, Mosier JA, McArdle TJ, Wang W, Schunk C, Jayathilake AL, Bates ME, Bordeleau F, Antonyak MA, Cerione RA, Reinhart-King CA. Weakly migratory metastatic breast cancer cells activate fibroblasts via microvesicle-Tg2 to facilitate dissemination and metastasis. eLife 2022; 11:e74433. [PMID: 36475545 PMCID: PMC9767463 DOI: 10.7554/elife.74433] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/05/2022] [Indexed: 12/12/2022] Open
Abstract
Cancer cell migration is highly heterogeneous, and the migratory capability of cancer cells is thought to be an indicator of metastatic potential. It is becoming clear that a cancer cell does not have to be inherently migratory to metastasize, with weakly migratory cancer cells often found to be highly metastatic. However, the mechanism through which weakly migratory cells escape from the primary tumor remains unclear. Here, utilizing phenotypically sorted highly and weakly migratory human breast cancer cells, we demonstrate that weakly migratory metastatic cells disseminate from the primary tumor via communication with stromal cells. While highly migratory cells are capable of single cell migration, weakly migratory cells rely on cell-cell signaling with fibroblasts to escape the primary tumor. Weakly migratory cells release microvesicles rich in tissue transglutaminase 2 (Tg2) which activate murine fibroblasts and lead weakly migratory cancer cell migration in vitro. These microvesicles also induce tumor stiffening and fibroblast activation in vivo and enhance the metastasis of weakly migratory cells. Our results identify microvesicles and Tg2 as potential therapeutic targets for metastasis and reveal a novel aspect of the metastatic cascade in which weakly migratory cells release microvesicles which activate fibroblasts to enhance cancer cell dissemination.
Collapse
Affiliation(s)
- Samantha C Schwager
- Department of Biomedical Engineering, Vanderbilt UniversityNashvilleUnited States
| | - Katherine M Young
- Department of Biomedical Engineering, Vanderbilt UniversityNashvilleUnited States
| | - Lauren A Hapach
- Department of Biomedical Engineering, Vanderbilt UniversityNashvilleUnited States
- Department of Biomedical Engineering, Cornell UniversityIthacaUnited States
| | - Caroline M Carlson
- Department of Biomedical Engineering, Vanderbilt UniversityNashvilleUnited States
| | - Jenna A Mosier
- Department of Biomedical Engineering, Vanderbilt UniversityNashvilleUnited States
| | | | - Wenjun Wang
- Department of Biomedical Engineering, Vanderbilt UniversityNashvilleUnited States
| | - Curtis Schunk
- Department of Biomedical Engineering, Vanderbilt UniversityNashvilleUnited States
| | | | - Madison E Bates
- Department of Biomedical Engineering, Vanderbilt UniversityNashvilleUnited States
| | - Francois Bordeleau
- CHU de Québec-Université Laval Research Center (Oncology division), UniversitéLaval Cancer Research Center and Faculty of Medicine, Université LavalQuébeccCanada
| | - Marc A Antonyak
- Department of Biomedical Science, Cornell UniversityIthacaUnited States
| | - Richard A Cerione
- Department of Biomedical Science, Cornell UniversityIthacaUnited States
| | | |
Collapse
|
16
|
TGFB2-AS1 inhibits triple-negative breast cancer progression via interaction with SMARCA4 and regulating its targets TGFB2 and SOX2. Proc Natl Acad Sci U S A 2022; 119:e2117988119. [PMID: 36126099 PMCID: PMC9522332 DOI: 10.1073/pnas.2117988119] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The multisubunit ATPase-dependent SWI/SNF complex plays an important role in chromatin remodeling. Large numbers of SWI/SNF subunit mutations have been identified in large variety of human cancers, suggesting that they function against tumorigenesis. Here we report long noncoding RNA TGFB2-AS1 correlates with prognosis in triple-negative breast cancer, the most aggressive cluster of all breast cancers. Especially, we show that TGFB2-AS1 interacts with SMARCA4, a core subunit of the SWI/SNF complex, and blocks the complex to approach its target promoters both in cis and in trans, thus inhibiting the expression of the target genes, TGFB2 and SOX2, eventually leading to the inhibition of breast cancer progression. These findings shed light on understanding regulation and roles of the SWI/SNF complex in carcinogenesis. Triple-negative breast cancer (TNBC) is the most challenging breast cancer subtype for its high rates of relapse, great metastatic potential, and short overall survival. How cancer cells acquire metastatic potency through the conversion of noncancer stem-like cells into cancer cells with stem-cell properties is poorly understood. Here, we identified the long noncoding RNA (lncRNA) TGFB2-AS1 as an important regulator of the reversibility and plasticity of noncancer stem cell populations in TNBC. We revealed that TGFB2-AS1 impairs the breast cancer stem-like cell (BCSC) traits of TNBC cells in vitro and dramatically decreases tumorigenic frequency and lung metastasis in vivo. Mechanistically, TGFB2-AS1 interacts with SMARCA4, a core subunit of the SWI/SNF chromatin remodeling complex, and results in transcriptional repression of its target genes including TGFB2 and SOX2 in an in cis or in trans way, leading to inhibition of transforming growth factor β (TGFβ) signaling and BCSC characteristics. In line with this, TGFB2-AS1 overexpression in an orthotopic TNBC mouse model remarkably abrogates the enhancement of tumor growth and lung metastasis endowed by TGFβ2. Furthermore, combined prognosis analysis of TGFB2-AS1 and TGFβ2 in TNBC patients shows that high TGFB2-AS1 and low TGFβ2 levels are correlated with better outcome. These findings demonstrate a key role of TGFB2-AS1 in inhibiting disease progression of TNBC based on switching the cancer cell fate of TNBC and also shed light on the treatment of TNBC patients.
Collapse
|
17
|
Yuan Y, Wang L, Zhao X, Wang J, Zhang M, Ma Q, Wei S, Yan Z, Cheng Y, Chen X, Zou H, Ge J, Wang Y, Zhang X, Cui Y, Luo T, Bian X. The E3 ubiquitin ligase HUWE1 acts through the N-Myc-DLL1-NOTCH1 signaling axis to suppress glioblastoma progression. Cancer Commun (Lond) 2022; 42:868-886. [PMID: 35848447 PMCID: PMC9456703 DOI: 10.1002/cac2.12334] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/21/2022] [Accepted: 07/03/2022] [Indexed: 06/15/2023] Open
Abstract
BACKGROUND Elucidation of the post-transcriptional modification has led to novel strategies to treat intractable tumors, especially glioblastoma (GBM). The ubiquitin-proteasome system (UPS) mediates a reversible, stringent and stepwise post-translational modification which is closely associated with malignant processes of GBM. To this end, developing novel therapeutic approaches to target the UPS may contribute to the treatment of this disease. This study aimed to screen the vital and aberrantly regulated component of the UPS in GBM. Based on the molecular identification, functional characterization, and mechanism investigation, we sought to elaborate a novel therapeutic strategy to target this vital factor to combat GBM. METHODS We combined glioma datasets and human patient samples to screen and identify aberrantly regulated E3 ubiquitin ligase. Multidimensional database analysis and molecular and functional experiments in vivo and in vitro were used to evaluate the roles of HECT, UBA and WWE domain-containing E3 ubiquitin ligase 1 (HUWE1) in GBM. dCas9 synergistic activation mediator system and recombinant adeno-associated virus (rAAV) were used to endogenously overexpress full-length HUWE1 in vitro and in glioma orthotopic xenografts. RESULTS Low expression of HUWE1 was closely associated with worse prognosis of GBM patients. The ubiquitination and subsequent degradation of N-Myc mediated by HUWE1, leading to the inactivation of downstream Delta-like 1 (DLL1)-NOTCH1 signaling pathways, inhibited the proliferation, invasion, and migration of GBM cells in vitro and in vivo. A rAAV dual-vector system for packaging and delivery of dCas9-VP64 was used to augment endogenous HUWE1 expression in vivo and showed an antitumor activity in glioma orthotopic xenografts. CONCLUSIONS The E3 ubiquitin ligase HUWE1 acts through the N-Myc-DLL1-NOTCH1 signaling axis to suppress GBM progression. Antitumor activity of rAAV dual-vector delivering dCas9-HUWE1 system uncovers a promising therapeutic strategy for GBM.
Collapse
Affiliation(s)
- Ye Yuan
- Institute of Pathology and Southwest Cancer CenterSouthwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of ChinaChongqing400038P. R. China
| | - Li‐Hong Wang
- Institute of Pathology and Southwest Cancer CenterSouthwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of ChinaChongqing400038P. R. China
| | - Xian‐Xian Zhao
- Department of Clinical LaboratorySouthwest HospitalThird Military Medical University (Army Medical University)Chongqing400038P. R. China
| | - Jiao Wang
- Institute of Pathology and Southwest Cancer CenterSouthwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of ChinaChongqing400038P. R. China
| | - Meng‐Si Zhang
- Institute of Pathology and Southwest Cancer CenterSouthwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of ChinaChongqing400038P. R. China
| | - Qing‐Hua Ma
- Institute of Pathology and Southwest Cancer CenterSouthwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of ChinaChongqing400038P. R. China
| | - Sen Wei
- Institute of Pathology and Southwest Cancer CenterSouthwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of ChinaChongqing400038P. R. China
| | - Ze‐Xuan Yan
- Institute of Pathology and Southwest Cancer CenterSouthwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of ChinaChongqing400038P. R. China
| | - Yue Cheng
- Institute of Pathology and Southwest Cancer CenterSouthwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of ChinaChongqing400038P. R. China
| | - Xiao‐Qing Chen
- Institute of Pathology and Southwest Cancer CenterSouthwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of ChinaChongqing400038P. R. China
| | - Hong‐Bo Zou
- Department of Oncologythe Third Affiliated Hospital of Chongqing Medical UniversityChongqing401120P. R. China
| | - Jia Ge
- Institute of Pathology and Southwest Cancer CenterSouthwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of ChinaChongqing400038P. R. China
| | - Yan Wang
- Institute of Pathology and Southwest Cancer CenterSouthwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of ChinaChongqing400038P. R. China
| | - Xia Zhang
- Institute of Pathology and Southwest Cancer CenterSouthwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of ChinaChongqing400038P. R. China
| | - You‐Hong Cui
- Institute of Pathology and Southwest Cancer CenterSouthwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of ChinaChongqing400038P. R. China
| | - Tao Luo
- Institute of Pathology and Southwest Cancer CenterSouthwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of ChinaChongqing400038P. R. China
| | - Xiu‐Wu Bian
- Institute of Pathology and Southwest Cancer CenterSouthwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of ChinaChongqing400038P. R. China
- Bio‐Bank of Southwest HospitalThird Military Medical University (Army Medical University)Chongqing400038P. R. China
| |
Collapse
|
18
|
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: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [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
|
19
|
Li XY, Pi YN, Chen Y, Zhu Q, Xia BR. Nicotinamide N-Methyltransferase: A Promising Biomarker and Target for Human Cancer Therapy. Front Oncol 2022; 12:894744. [PMID: 35756670 PMCID: PMC9218565 DOI: 10.3389/fonc.2022.894744] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 04/29/2022] [Indexed: 11/13/2022] Open
Abstract
Cancer cells typically exhibit a tightly regulated program of metabolic plasticity and epigenetic remodeling to meet the demand of uncontrolled cell proliferation. The metabolic-epigenetic axis has recently become an increasingly hot topic in carcinogenesis and offers new avenues for innovative and personalized cancer treatment strategies. Nicotinamide N-methyltransferase (NNMT) is a metabolic enzyme involved in controlling methylation potential, impacting DNA and histone epigenetic modification. NNMT overexpression has been described in various solid cancer tissues and even body fluids, including serum, urine, and saliva. Furthermore, accumulating evidence has shown that NNMT knockdown significantly decreases tumorigenesis and chemoresistance capacity. Most importantly, the natural NNMT inhibitor yuanhuadine can reverse epidermal growth factor receptor tyrosine kinase inhibitor resistance in lung cancer cells. In this review, we evaluate the possibility of NNMT as a diagnostic biomarker and molecular target for effective anticancer treatment. We also reveal the exact mechanisms of how NNMT affects epigenetics and the development of more potent and selective inhibitors.
Collapse
Affiliation(s)
- Xiao-Yu Li
- The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Ya-Nan Pi
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yao Chen
- Department of Gynecology, Bengbu Medical College Bengbu, Anhui, China
| | - Qi Zhu
- The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Bai-Rong Xia
- The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Provincial Cancer Hospital, Hefei, China
| |
Collapse
|
20
|
Reustle A, Menig LS, Leuthold P, Hofmann U, Stühler V, Schmees C, Becker M, Haag M, Klumpp V, Winter S, Büttner FA, Rausch S, Hennenlotter J, Fend F, Scharpf M, Stenzl A, Bedke J, Schwab M, Schaeffeler E. Nicotinamide-N-methyltransferase is a promising metabolic drug target for primary and metastatic clear cell renal cell carcinoma. Clin Transl Med 2022; 12:e883. [PMID: 35678045 PMCID: PMC9178377 DOI: 10.1002/ctm2.883] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 04/27/2022] [Accepted: 05/03/2022] [Indexed: 12/16/2022] Open
Abstract
Background The metabolic enzyme nicotinamide‐N‐methyltransferase (NNMT) is highly expressed in various cancer entities, suggesting tumour‐promoting functions. We systematically investigated NNMT expression and its metabolic interactions in clear cell renal cell carcinoma (ccRCC), a prominent RCC subtype with metabolic alterations, to elucidate its role as a drug target. Methods NNMT expression was assessed in primary ccRCC (n = 134), non‐tumour tissue and ccRCC‐derived metastases (n = 145) by microarray analysis and/or immunohistochemistry. Findings were validated in The Cancer Genome Atlas (kidney renal clear cell carcinoma [KIRC], n = 452) and by single‐cell analysis. Expression was correlated with clinicopathological data and survival. Metabolic alterations in NNMT‐depleted cells were assessed by nontargeted/targeted metabolomics and extracellular flux analysis. The NNMT inhibitor (NNMTi) alone and in combination with the inhibitor 2‐deoxy‐D‐glucose for glycolysis and BPTES (bis‐2‐(5‐phenylacetamido‐1,3,4‐thiadiazol‐2‐yl)ethyl‐sulfide) for glutamine metabolism was investigated in RCC cell lines (786‐O, A498) and in two 2D ccRCC‐derived primary cultures and three 3D ccRCC air–liquid interface models. Results NNMT protein was overexpressed in primary ccRCC (p = 1.32 × 10–16) and ccRCC‐derived metastases (p = 3.92 × 10–20), irrespective of metastatic location, versus non‐tumour tissue. Single‐cell data showed predominant NNMT expression in ccRCC and not in the tumour microenvironment. High NNMT expression in primary ccRCC correlated with worse survival in independent cohorts (primary RCC—hazard ratio [HR] = 4.3, 95% confidence interval [CI]: 1.5–12.4; KIRC—HR = 3.3, 95% CI: 2.0–5.4). NNMT depletion leads to intracellular glutamine accumulation, with negative effects on mitochondrial function and cell survival, while not affecting glycolysis or glutathione metabolism. At the gene level, NNMT‐depleted cells upregulate glycolysis, oxidative phosphorylation and apoptosis pathways. NNMTi alone or in combination with 2‐deoxy‐D‐glucose and BPTES resulted in inhibition of cell viability in ccRCC cell lines and primary tumour and metastasis‐derived models. In two out of three patient‐derived ccRCC air–liquid interface models, NNMTi treatment induced cytotoxicity. Conclusions Since efficient glutamine utilisation, which is essential for ccRCC tumours, depends on NNMT, small‐molecule NNMT inhibitors provide a novel therapeutic strategy for ccRCC and act as sensitizers for combination therapies.
Collapse
Affiliation(s)
- Anna Reustle
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,University of Tuebingen, Tuebingen, Germany
| | - Lena-Sophie Menig
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,University of Tuebingen, Tuebingen, Germany
| | - Patrick Leuthold
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,University of Tuebingen, Tuebingen, Germany
| | - Ute Hofmann
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,University of Tuebingen, Tuebingen, Germany
| | - Viktoria Stühler
- Department of Urology, University Hospital Tuebingen, Tuebingen, Germany
| | - Christian Schmees
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, Reutlingen, Germany
| | - Michael Becker
- Experimental Pharmacology and Oncology GmbH, Berlin-Buch, Germany
| | - Mathias Haag
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,University of Tuebingen, Tuebingen, Germany
| | - Verena Klumpp
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,University of Tuebingen, Tuebingen, Germany
| | - Stefan Winter
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,University of Tuebingen, Tuebingen, Germany
| | - Florian A Büttner
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,University of Tuebingen, Tuebingen, Germany
| | - Steffen Rausch
- Department of Urology, University Hospital Tuebingen, Tuebingen, Germany
| | - Jörg Hennenlotter
- Department of Urology, University Hospital Tuebingen, Tuebingen, Germany
| | - Falko Fend
- Institute of Pathology and Neuropathology, University Hospital Tuebingen, Tuebingen, Germany
| | - Marcus Scharpf
- Institute of Pathology and Neuropathology, University Hospital Tuebingen, Tuebingen, Germany
| | - Arnulf Stenzl
- Department of Urology, University Hospital Tuebingen, Tuebingen, Germany
| | - Jens Bedke
- Department of Urology, University Hospital Tuebingen, Tuebingen, Germany.,German Cancer Consortium (DKTK), Partner Site Tübingen, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,University of Tuebingen, Tuebingen, Germany.,German Cancer Consortium (DKTK), Partner Site Tübingen, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Departments of Clinical Pharmacology, Pharmacy and Biochemistry, University of Tuebingen, Tuebingen, Germany.,Cluster of Excellence iFIT (EXC2180) 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tuebingen, Tuebingen, Germany
| | - Elke Schaeffeler
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,University of Tuebingen, Tuebingen, Germany.,Cluster of Excellence iFIT (EXC2180) 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
21
|
Fan SY, Long SY, Liu JJ, Zhang WL, Hu JL. Nicotinamide N-Methyltransferase inhibits HBV replication by suppressing NR5A1 expression invitro. Biochem Biophys Res Commun 2022; 614:70-77. [PMID: 35569378 DOI: 10.1016/j.bbrc.2022.04.122] [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: 04/19/2022] [Accepted: 04/26/2022] [Indexed: 11/02/2022]
Abstract
Chronic hepatitis B virus (HBV) infection can lead to fibrosis, liver cirrhosis, and primary hepatocellular carcinoma. Investigating host factors that regulate HBV replication helps to identify antiviral targets. In the current study, we identified Nicotinamide N-Methyltransferase gene (NNMT) as a novel factor that regulates HBV transcription. NNMT is up-regulated at both the mRNA and protein levels in HepG2.2.15 cells compared to HepG2 cells. Overexpression of NNMT reduces HBV replication in several cell models, while knockdown of NNMT enhances HBV DNA levels. Mechanistically, NNMT suppresses HBV DNA replication by inhibiting HBV RNA transcription. The region required for the inhibitory effect of NNMT was narrowed to nt 1672-1708 in enhancer II by luciferase assays. On the other hand, ChIP assays and EMSA results showed that NNMT does not bind to this region substantially, either directly or indirectly. Next, a collection of hepatic nuclear receptor transcription factors was screened to determine whether they were affected by NNMT overexpression. NR5A1, a positive regulator of HBV replication, decreased significantly after NNMT overexpression. Collectively, the findings of this study shed light on the regulation of HBV transcription.
Collapse
Affiliation(s)
- Shu-Ying Fan
- Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Shao-Yuan Long
- Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Jia-Jun Liu
- Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Wen-Lu Zhang
- Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing, China.
| | - Jie-Li Hu
- Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing, China
| |
Collapse
|
22
|
A proteogenomic analysis of clear cell renal cell carcinoma in a Chinese population. Nat Commun 2022; 13:2052. [PMID: 35440542 PMCID: PMC9019091 DOI: 10.1038/s41467-022-29577-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 02/25/2022] [Indexed: 12/16/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is a common and aggressive subtype of renal cancer. Here we conduct a comprehensive proteogenomic analysis of 232 tumor and adjacent non-tumor tissue pairs from Chinese ccRCC patients. By comparing with tumor adjacent tissues, we find that ccRCC shows extensive metabolic dysregulation and an enhanced immune response. Molecular subtyping classifies ccRCC tumors into three subtypes (GP1–3), among which the most aggressive GP1 exhibits the strongest immune phenotype, increased metastasis, and metabolic imbalance, linking the multi-omics-derived phenotypes to clinical outcomes of ccRCC. Nicotinamide N-methyltransferase (NNMT), a one-carbon metabolic enzyme, is identified as a potential marker of ccRCC and a drug target for GP1. We demonstrate that NNMT induces DNA-dependent protein kinase catalytic subunit (DNA-PKcs) homocysteinylation, increases DNA repair, and promotes ccRCC tumor growth. This study provides insights into the biological underpinnings and prognosis assessment of ccRCC, revealing targetable metabolic vulnerabilities. Clear cell renal cell carcinoma is an aggressive form of renal cancer, with differences in genomic mutations reported between Western and Eastern populations. In this study, the authors have compiled proteogenomic analysis of Chinese ccRCC to reveal genomic alterations and dysregulation of immune and metabolic responses.
Collapse
|
23
|
Wang W, Yang C, Wang T, Deng H. Complex roles of nicotinamide N-methyltransferase in cancer progression. Cell Death Dis 2022; 13:267. [PMID: 35338115 PMCID: PMC8956669 DOI: 10.1038/s41419-022-04713-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/23/2022] [Accepted: 03/08/2022] [Indexed: 02/07/2023]
Abstract
Nicotinamide N-methyltransferase (NNMT) is an intracellular methyltransferase, catalyzing the N-methylation of nicotinamide (NAM) to form 1-methylnicotinamide (1-MNAM), in which S-adenosyl-l-methionine (SAM) is the methyl donor. High expression of NNMT can alter cellular NAM and SAM levels, which in turn, affects nicotinamide adenine dinucleotide (NAD+)-dependent redox reactions and signaling pathways, and remodels cellular epigenetic states. Studies have revealed that NNMT plays critical roles in the occurrence and development of various cancers, and analysis of NNMT expression levels in different cancers from The Cancer Genome Atlas (TCGA) dataset indicated that NNMT might be a potential biomarker and therapeutic target for tumor diagnosis and treatment. This review provides a comprehensive understanding of recent advances on NNMT functions in different tumors and deciphers the complex roles of NNMT in cancer progression.
Collapse
Affiliation(s)
- Weixuan Wang
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, People's Republic of China
| | - Changmei Yang
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing, People's Republic of China
| | - Tianxiang Wang
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing, People's Republic of China
| | - Haiteng Deng
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing, People's Republic of China.
| |
Collapse
|
24
|
Das D, Karthik N, Taneja R. Epigenetic Small-Molecule Modulators Targeting Metabolic Pathways in Cancer. Subcell Biochem 2022; 100:523-555. [PMID: 36301505 DOI: 10.1007/978-3-031-07634-3_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Metabolic deregulation is a key factor in cancer progression. Epigenetic changes and metabolic rewiring are intertwined in cancer. Deregulated epigenetic modifiers cause metabolic aberrations by targeting the expression of metabolic enzymes. Conversely, metabolites and cofactors affect the expression and activity of epigenetic regulators. Small molecules are promising therapeutic approaches to target the epigenetic-metabolomic crosstalk in cancer. Here, we focus on the interplay between metabolic rewiring and epigenetic landscape in the context of tumourigenesis and highlight recent advances in the use of small-molecule drug targets for therapy.
Collapse
Affiliation(s)
- Dipanwita Das
- Department of Physiology and Healthy Longevity Translational Research Program Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Nandini Karthik
- Department of Physiology and Healthy Longevity Translational Research Program Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Reshma Taneja
- Department of Physiology and Healthy Longevity Translational Research Program Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
25
|
Togni L, Mascitti M, Sartini D, Campagna R, Pozzi V, Salvolini E, Offidani A, Santarelli A, Emanuelli M. Nicotinamide N-Methyltransferase in Head and Neck Tumors: A Comprehensive Review. Biomolecules 2021; 11:1594. [PMID: 34827592 PMCID: PMC8615955 DOI: 10.3390/biom11111594] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 10/19/2021] [Accepted: 10/26/2021] [Indexed: 12/31/2022] Open
Abstract
The head and neck tumors (HNT) are a heterogeneous group of diseases ranging from benign to malignant lesions, with distinctive molecular and clinical behaviors. Several studies have highlighted the presence of an altered metabolic phenotype in HNT, such as the upregulation of nicotinamide N-methyltransferase (NNMT). However, its biological effects have not been completely disclosed and the role of NNMT in cancer cell metabolism remains unclear. Therefore, this comprehensive review aims to evaluate the available literature regarding the biological, diagnostic, and prognostic role of NNMT in HNT. NNMT was shown to be significantly overexpressed in all of the evaluated HNT types. Moreover, its upregulation has been correlated with cancer cell migration and adverse clinical outcomes, such as high-pathological stage, lymph node metastasis, and locoregional recurrences. However, in oral squamous cell carcinoma (OSCC) these associations are still debated, and several studies have failed to demonstrate the prognostic significance of NNMT. The shRNA-mediated gene silencing efficiently suppressed the NNMT gene expression and exhibited a clear inhibitory effect on cell proliferation, promoting the expression of apoptosis-related proteins and modulating the cell cycle. NNMT could represent a new molecular biomarker and a new target of molecular-based therapy, although further studies on larger patient cohorts are needed to explore its biological role in HNT.
Collapse
Affiliation(s)
- Lucrezia Togni
- Department of Clinical, Specialistic and Dental Sciences, Marche Polytechnic University, 60126 Ancona, Italy; (L.T.); (M.M.); (D.S.); (R.C.); (V.P.); (E.S.); (M.E.)
| | - Marco Mascitti
- Department of Clinical, Specialistic and Dental Sciences, Marche Polytechnic University, 60126 Ancona, Italy; (L.T.); (M.M.); (D.S.); (R.C.); (V.P.); (E.S.); (M.E.)
| | - Davide Sartini
- Department of Clinical, Specialistic and Dental Sciences, Marche Polytechnic University, 60126 Ancona, Italy; (L.T.); (M.M.); (D.S.); (R.C.); (V.P.); (E.S.); (M.E.)
| | - Roberto Campagna
- Department of Clinical, Specialistic and Dental Sciences, Marche Polytechnic University, 60126 Ancona, Italy; (L.T.); (M.M.); (D.S.); (R.C.); (V.P.); (E.S.); (M.E.)
| | - Valentina Pozzi
- Department of Clinical, Specialistic and Dental Sciences, Marche Polytechnic University, 60126 Ancona, Italy; (L.T.); (M.M.); (D.S.); (R.C.); (V.P.); (E.S.); (M.E.)
| | - Eleonora Salvolini
- Department of Clinical, Specialistic and Dental Sciences, Marche Polytechnic University, 60126 Ancona, Italy; (L.T.); (M.M.); (D.S.); (R.C.); (V.P.); (E.S.); (M.E.)
| | - Annamaria Offidani
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, 60126 Ancona, Italy;
| | - Andrea Santarelli
- Department of Clinical, Specialistic and Dental Sciences, Marche Polytechnic University, 60126 Ancona, Italy; (L.T.); (M.M.); (D.S.); (R.C.); (V.P.); (E.S.); (M.E.)
- Dentistry Clinic, National Institute of Health and Science of Aging, IRCCS INRCA, 60126 Ancona, Italy
| | - Monica Emanuelli
- Department of Clinical, Specialistic and Dental Sciences, Marche Polytechnic University, 60126 Ancona, Italy; (L.T.); (M.M.); (D.S.); (R.C.); (V.P.); (E.S.); (M.E.)
| |
Collapse
|
26
|
Tatekawa S, Ofusa K, Chijimatsu R, Vecchione A, Tamari K, Ogawa K, Ishii H. Methylosystem for Cancer Sieging Strategy. Cancers (Basel) 2021; 13:5088. [PMID: 34680237 PMCID: PMC8534198 DOI: 10.3390/cancers13205088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 10/02/2021] [Accepted: 10/07/2021] [Indexed: 12/14/2022] Open
Abstract
As cancer is a genetic disease, methylation defines a biologically malignant phenotype of cancer in the association of one-carbon metabolism-dependent S-adenosylmethionine (SAM) as a methyl donor in each cell. Methylated substances are involved in intracellular metabolism, but via intercellular communication, some of these can also be secreted to affect other substances. Although metabolic analysis at the single-cell level remains challenging, studying the "methylosystem" (i.e., the intercellular and intracellular communications of upstream regulatory factors and/or downstream effectors that affect the epigenetic mechanism involving the transfer of a methyl group from SAM onto the specific positions of nucleotides or other metabolites in the tumor microenvironment) and tracking these metabolic products are important research tasks for understanding spatial heterogeneity. Here, we discuss and highlight the involvement of RNA and nicotinamide, recently emerged targets, in SAM-producing one-carbon metabolism in cancer cells, cancer-associated fibroblasts, and immune cells. Their significance and implications will contribute to the discovery of efficient methods for the diagnosis of and therapeutic approaches to human cancer.
Collapse
Affiliation(s)
- Shotaro Tatekawa
- Department of Radiation Oncology, Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (S.T.); (K.T.)
| | - Ken Ofusa
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (K.O.); (R.C.)
- Food and Life-Science Laboratory, Prophoenix Division, Idea Consultants, Inc., Osaka 559-8519, Japan
| | - Ryota Chijimatsu
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (K.O.); (R.C.)
| | - Andrea Vecchione
- Department of Clinical and Molecular Medicine, University of Rome “Sapienza”, Santo Andrea Hospital, Via di Grottarossa, 1035-00189 Rome, Italy;
| | - Keisuke Tamari
- Department of Radiation Oncology, Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (S.T.); (K.T.)
| | - Kazuhiko Ogawa
- Department of Radiation Oncology, Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (S.T.); (K.T.)
| | - Hideshi Ishii
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (K.O.); (R.C.)
| |
Collapse
|
27
|
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: 8.0] [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
|
28
|
Roles of Nicotinamide N-Methyltransferase in Obesity and Type 2 Diabetes. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9924314. [PMID: 34368359 PMCID: PMC8337113 DOI: 10.1155/2021/9924314] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 07/20/2021] [Indexed: 11/28/2022]
Abstract
Type 2 diabetes (T2D) is thought to be a complication of metabolic syndrome caused by disorders of energy utilization and storage and characterized by insulin resistance or deficiency of insulin secretion. Though the mechanism linking obesity to the development of T2D is complex and unintelligible, it is known that abnormal lipid metabolism and adipose tissue accumulation possibly play important roles in this process. Recently, nicotinamide N-methyltransferase (NNMT) has been emerging as a new mechanism-of-action target in treating obesity and associated T2D. Evidence has shown that NNMT is associated with obesity and T2D. NNMT inhibition or NNMT knockdown significantly increases energy expenditure, reduces body weight and white adipose mass, improves insulin sensitivity, and normalizes glucose tolerance and fasting blood glucose levels. Additionally, trials of oligonucleotide therapeutics and experiments with some small-molecule NNMT inhibitors in vitro and in preclinical animal models have validated NNMT as a promising therapeutic target to prevent or treat obesity and associated T2D. However, the exact mechanisms underlying these phenomena are not yet fully understood and clinical trials targeting NNMT have not been reported until now. Therefore, more researches are necessary to reveal the acting mechanism of NNMT in obesity and T2D and to develop therapeutics targeting NNMT.
Collapse
|
29
|
Aldughaim MS, Al-Anazi MR, Bohol MFF, Colak D, Alothaid H, Wakil SM, Hagos ST, Ali D, Alarifi S, Rout S, Alkahtani S, Al-Ahdal MN, Al-Qahtani AA. Gene Expression and Transcriptome Profiling of Changes in a Cancer Cell Line Post-Exposure to Cadmium Telluride Quantum Dots: Possible Implications in Oncogenesis. Dose Response 2021; 19:15593258211019880. [PMID: 34177396 PMCID: PMC8202281 DOI: 10.1177/15593258211019880] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 04/28/2021] [Accepted: 04/30/2021] [Indexed: 11/29/2022] Open
Abstract
Cadmium telluride quantum dots (CdTe-QDs) are acquiring great interest in terms of their applications in biomedical sciences. Despite earlier sporadic studies on possible oncogenic roles and anticancer properties of CdTe-QDs, there is limited information regarding the oncogenic potential of CdTe-QDs in cancer progression. Here, we investigated the oncogenic effects of CdTe-QDs on the gene expression profiles of Chang cancer cells. Chang cancer cells were treated with 2 different doses of CdTe-QDs (10 and 25 μg/ml) at different time intervals (6, 12, and 24 h). Functional annotations helped identify the gene expression profile in terms of its biological process, canonical pathways, and gene interaction networks activated. It was found that the gene expression profiles varied in a time and dose-dependent manner. Validation of transcriptional changes of several genes through quantitative PCR showed that several genes upregulated by CdTe-QD exposure were somewhat linked with oncogenesis. CdTe-QD-triggered functional pathways that appear to associate with gene expression, cell proliferation, migration, adhesion, cell-cycle progression, signal transduction, and metabolism. Overall, CdTe-QD exposure led to changes in the gene expression profiles of the Chang cancer cells, highlighting that this nanoparticle can further drive oncogenesis and cancer progression, a finding that indicates the merit of immediate in vivo investigation.
Collapse
Affiliation(s)
| | - Mashael R Al-Anazi
- Department of Infection and Immunity, Research Centre, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Marie Fe F Bohol
- Department of Infection and Immunity, Research Centre, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Dilek Colak
- Department of Biostatistics, Epidemiology and Scientific Computing, Research Centre, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Hani Alothaid
- Department of Basic Medical Sciences, Faculty of Applied Medical Sciences, Al-Baha University, Al-Baha, Saudi Arabia
| | - Salma Majid Wakil
- Genotyping Core Facility, Research Centre, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Samya T Hagos
- Genotyping Core Facility, Research Centre, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Daoud Ali
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Saud Alarifi
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Sashmita Rout
- Advanced Centre for Treatment, Research, and Education in Cancer, Tata memorial Hospital, Mumbai, India
| | - Saad Alkahtani
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Mohammed N Al-Ahdal
- Department of Infection and Immunity, Research Centre, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia.,Department of Microbiology and Immunology, Alfaisal University, School of Medicine, Riyadh, Saudi Arabia
| | - Ahmed A Al-Qahtani
- Department of Infection and Immunity, Research Centre, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia.,Department of Microbiology and Immunology, Alfaisal University, School of Medicine, Riyadh, Saudi Arabia
| |
Collapse
|
30
|
Campagna R, Salvolini E, Pompei V, Pozzi V, Salvucci A, Molinelli E, Brisigotti V, Sartini D, Campanati A, Offidani A, Emanuelli M. Nicotinamide N-methyltransferase gene silencing enhances chemosensitivity of melanoma cell lines. Pigment Cell Melanoma Res 2021; 34:1039-1048. [PMID: 34018676 DOI: 10.1111/pcmr.12993] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/11/2021] [Accepted: 05/17/2021] [Indexed: 11/28/2022]
Abstract
Melanoma accounts for less than 5% of all cutaneous neoplasms but is responsible for the greater part of skin cancer-related deaths. Therefore, the identification of molecules that could serve as the therapeutic target is urgent. This study focused on the enzyme nicotinamide N-methyltransferase (NNMT). The effect of NNMT knockdown on cell proliferation and migration of A375 melanoma cells was evaluated by MTT and wound healing assays, respectively. Viability of A375 cells downregulating NNMT was also explored under treatment with dacarbazine, a chemotherapeutic drug approved for advanced melanoma treatment. The impact of enzyme knockdown on cell proliferation and chemosensitivity was also investigated in WM-115 melanoma cells. Results obtained demonstrated that NNMT silencing led to a significant reduction of cell proliferation and migration of A375 cells. Moreover, enzyme downregulation was associated with an increase of melanoma cells sensitivity to treatment with dacarbazine. Analogous effects induced by enzyme knockdown on cell proliferation and chemosensitivity were also found in the WM-115 cell line. Our data seem to demonstrate that NNMT could represent a promising molecular target for the effective treatment of this form of skin cancer.
Collapse
Affiliation(s)
- Roberto Campagna
- Department of Clinical Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Eleonora Salvolini
- Department of Clinical Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Veronica Pompei
- Department of Clinical Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Valentina Pozzi
- Department of Clinical Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Alessia Salvucci
- Department of Clinical 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
| | - 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
|
31
|
Hapach LA, Carey SP, Schwager SC, Taufalele PV, Wang W, Mosier JA, Ortiz-Otero N, McArdle TJ, Goldblatt ZE, Lampi MC, Bordeleau F, Marshall JR, Richardson IM, Li J, King MR, Reinhart-King CA. Phenotypic Heterogeneity and Metastasis of Breast Cancer Cells. Cancer Res 2021; 81:3649-3663. [PMID: 33975882 DOI: 10.1158/0008-5472.can-20-1799] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 02/02/2021] [Accepted: 05/05/2021] [Indexed: 11/16/2022]
Abstract
Although intratumoral genomic heterogeneity can impede cancer research and treatment, less is known about the effects of phenotypic heterogeneities. To investigate the role of cell migration heterogeneities in metastasis, we phenotypically sorted metastatic breast cancer cells into two subpopulations based on migration ability. Although migration is typically considered to be associated with metastasis, when injected orthotopically in vivo, the weakly migratory subpopulation metastasized significantly more than the highly migratory subpopulation. To investigate the mechanism behind this observation, both subpopulations were assessed at each stage of the metastatic cascade, including dissemination from the primary tumor, survival in the circulation, extravasation, and colonization. Although both subpopulations performed each step successfully, weakly migratory cells presented as circulating tumor cell (CTC) clusters in the circulation, suggesting clustering as one potential mechanism behind the increased metastasis of weakly migratory cells. RNA sequencing revealed weakly migratory subpopulations to be more epithelial and highly migratory subpopulations to be more mesenchymal. Depletion of E-cadherin expression from weakly migratory cells abrogated metastasis. Conversely, induction of E-cadherin expression in highly migratory cells increased metastasis. Clinical patient data and blood samples showed that CTC clustering and E-cadherin expression are both associated with worsened patient outcome. This study demonstrates that deconvolving phenotypic heterogeneities can reveal fundamental insights into metastatic progression. More specifically, these results indicate that migratory ability does not necessarily correlate with metastatic potential and that E-cadherin promotes metastasis in phenotypically sorted breast cancer cell subpopulations by enabling CTC clustering. SIGNIFICANCE: This study employs phenotypic cell sorting for migration to reveal a weakly migratory, highly metastatic breast cancer cell subpopulation regulated by E-cadherin, highlighting the dichotomy between cancer cell migration and metastasis.
Collapse
Affiliation(s)
- Lauren A Hapach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York.,Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Shawn P Carey
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York
| | - Samantha C Schwager
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Paul V Taufalele
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Wenjun Wang
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Jenna A Mosier
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Nerymar Ortiz-Otero
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York
| | | | - Zachary E Goldblatt
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York
| | - Marsha C Lampi
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York
| | - Francois Bordeleau
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee.,CHU de Québec-Université Laval Research Center, Université Laval Cancer Research Center, Québec, Canada
| | - Jocelyn R Marshall
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York
| | - Isaac M Richardson
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Jiahe Li
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York
| | - Michael R King
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | | |
Collapse
|
32
|
Li J, Yue H, Yu H, Lu X, Xue X. Patients with low nicotinamide N-methyltransferase expression benefit significantly from bevacizumab treatment in ovarian cancer. BMC Cancer 2021; 21:67. [PMID: 33446144 PMCID: PMC7809740 DOI: 10.1186/s12885-021-07785-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 01/02/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND The role of nicotinamide N-methyltransferase (NNMT) in ovarian cancer is still elusive. Our aim is to explore the expression of NNMT in ovarian cancer and to assess its association with patient prognosis and treatment response. METHODS We first analyzed the differential expression of NNMT among fallopian tube epithelium, primary ovarian cancers, metastatic ovarian cancers, and recurrent ovarian cancers using Gene Expression Ominus (GEO) database (GSE10971, GSE30587, GSE44104 and TCGA datasets). Then, we assessed the association of NNMT expression with clinical and molecular parameters using CSIOVDB database and GSE28739 dataset. Next, we evaluate the association of NNMT expression with the prognosis of ovarian cancer patients in both GSE9891 dataset and TCGA dataset. Finally, GSE140082 dataset was used to explore the association of NNMT expression with bevacizumab response. RESULTS NNMT expression was significantly elevated in lymphovascular space invasion (LVSI)-positive ovarian cancers compared with that in LVSI-negative ovarian cancers (TCGA dataset, P < 0.05), Moreover, increased expression of NNMT was associated with increased tumor stage, grade, and mesenchymal molecular subtype (CSIOVDB database). Survival analysis indicated that increased expression of NNMT was associated with a reduced OS in both GSE9891 dataset (HR: 2.28, 95%CI: 1.51-3.43, Log-rank P < 0.001) and TCGA dataset (HR: 1.55, 95%CI: 1.02-2.36, Log-rank P = 0.039). Multivariate analysis further confirmed the negative impact of NNMT expression on OS in ovarian cancer patients in those two datasets. Furthermore, the NNMT-related nomogram showed that NNMT shared a larger contribution to OS, compared with debulking status. More interestingly, bevacizumab conferred significant improvements in OS for patients with low NNMT expression (HR: 0.56, 95%CI: 0.31-0.99, Log-rank P = 0.049). In contrast, patients with high NNMT expression didn't benefit from bevacizumab treatment significantly (HR: 0.85, 95%CI: 0.48-1.49, Log-rank P = 0.561). NNMT expression was positively correlated with the expression of genes, LDHA and PGAM1, involved in Warburg effect. CONCLUSIONS In conclusion, NNMT expression is associated with the aggressive behavior of ovarian cancer, correlates with a poor prognosis, and is predictive of sensitivity to bevacizumab treatment.
Collapse
Affiliation(s)
- Jun Li
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, No.419, Fangxie Road, Shanghai, 200011, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, 200011, China
| | - Huiran Yue
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, No.419, Fangxie Road, Shanghai, 200011, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, 200011, China
| | - Hailin Yu
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, No.419, Fangxie Road, Shanghai, 200011, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, 200011, China
| | - Xin Lu
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, No.419, Fangxie Road, Shanghai, 200011, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, 200011, China
| | - Xiaohong Xue
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, No.419, Fangxie Road, Shanghai, 200011, China. .,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, 200011, China.
| |
Collapse
|
33
|
Nerve growth factor interacts with CHRM4 and promotes neuroendocrine differentiation of prostate cancer and castration resistance. Commun Biol 2021; 4:22. [PMID: 33398073 PMCID: PMC7782543 DOI: 10.1038/s42003-020-01549-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 12/01/2020] [Indexed: 02/06/2023] Open
Abstract
Nerve growth factor (NGF) contributes to the progression of malignancy. However, the functional role and regulatory mechanisms of NGF in the development of neuroendocrine prostate cancer (NEPC) are unclear. Here, we show that an androgen-deprivation therapy (ADT)-stimulated transcription factor, ZBTB46, upregulated NGF via ZBTB46 mediated-transcriptional activation of NGF. NGF regulates NEPC differentiation by physically interacting with a G-protein-coupled receptor, cholinergic receptor muscarinic 4 (CHRM4), after ADT. Pharmacologic NGF blockade and NGF knockdown markedly inhibited CHRM4-mediated NEPC differentiation and AKT-MYCN signaling activation. CHRM4 stimulation was associated with ADT resistance and was significantly correlated with increased NGF in high-grade and small-cell neuroendocrine prostate cancer (SCNC) patient samples. Our results reveal a role of the NGF in the development of NEPC that is linked to ZBTB46 upregulation and CHRM4 accumulation. Our study provides evidence that the NGF-CHRM4 axis has potential to be considered as a therapeutic target to impair NEPC progression. Here, the authors discover that NGF, upregulated by transcription factor ZBTB46 in prostate cancer exposed to androgen therapy, promotes neuroendocrine differentiation. They show that NGF interacts with the GPCR CHRM4, that both NGF and CHRM4 are upregulated in highly metastatic prostate cancer and that targeting NGF reduces therapy resistance in a mouse xenograft model.
Collapse
|
34
|
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]
|
35
|
Wu M, Hu W, Wang G, Yao Y, Yu XF. Nicotinamide N-Methyltransferase Is a Prognostic Biomarker and Correlated With Immune Infiltrates in Gastric Cancer. Front Genet 2020; 11:580299. [PMID: 33193702 PMCID: PMC7655872 DOI: 10.3389/fgene.2020.580299] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 09/10/2020] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer (GC) is the third most common cause of cancer-related death in the word. Immunotherapy is a promising treatment of cancer. However, it is unclear which GC subpopulation would benefit most from immunotherapy and it is necessary to develop effective biomarkers for predicting immunotherapy response. Nicotinamide N-methyltransferase (NNMT) is a metabolic regulator of cancer-associated fibroblast (CAF) differentiation and cancer progression. In this study, we explored the correlations of NNMT to tumor-infiltrating immune cells (TIICs) and immune marker sets in The Cancer Genome Atlas Stomach Adenocarcinoma STAD (TCGA-STAD). Subsequently, we screened the NNMT correlated genes and performed the enrichment analysis of these genes. We eventually predicted the 19 most potential small-molecule drugs using the connectivity map (CMap) and Comparative Toxicogenomics Database (CTD). Also, nadolol, tranexamic acid, felbinac and dapsone were considered the four most promising drugs for GC. In summary, NNMT can be used as a prognostic biomarker that reflect immune infiltration level and a novel therapeutic target in GC.
Collapse
Affiliation(s)
- Miaowei Wu
- Cancer Institute, Second Affiliated Hospital, Cancer Institute, Zhejiang University School of Medicine, Hangzhou, China
| | - Weilei Hu
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Guosheng Wang
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Yihan Yao
- Department of Surgical Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao-Fang Yu
- Cancer Institute, Second Affiliated Hospital, Cancer Institute, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
36
|
Li Y, Zhang Y, Cheng Y, Du T, Zhang J. Solvent inhibition profiles and inverse solvent isotope effects for enzymatic methyl transfer catalyzed by nicotinamide N‐methyltransferase. J PHYS ORG CHEM 2020. [DOI: 10.1002/poc.4093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Yuping Li
- School of Pharmaceutical Science and Technology Tianjin University Tianjin China
| | - Yali Zhang
- School of Pharmaceutical Science and Technology Tianjin University Tianjin China
| | - Yiting Cheng
- School of Pharmaceutical Science and Technology Tianjin University Tianjin China
| | - Tianshu Du
- School of Pharmaceutical Science and Technology Tianjin University Tianjin China
| | - Jianyu Zhang
- School of Pharmaceutical Science and Technology Tianjin University Tianjin China
| |
Collapse
|
37
|
Lakshmanan VK, Ojha S, Jung YD. A modern era of personalized medicine in the diagnosis, prognosis, and treatment of prostate cancer. Comput Biol Med 2020; 126:104020. [PMID: 33039808 DOI: 10.1016/j.compbiomed.2020.104020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/23/2020] [Accepted: 09/23/2020] [Indexed: 12/24/2022]
Abstract
The present era is witnessing rapid advancements in the field of medical informatics and modern healthcare management. The role of translational bioinformatics (TBI), an infant discipline in the field of medical informatics, is pivotal in this revolution. The development of high-throughput technologies [e.g., microarrays, next-generation sequencing (NGS)] has propelled TBI to the next stage in this modern era of medical informatics. In this review, we assess the promising translational outcomes of microarray- and NGS-based discovery of genes, proteins, micro RNAs, and other active biological compounds aiding in the diagnosis, prognosis, and therapy of prostate cancer (PCa) to improve treatment strategies at the localized and/or metastatic stages in patients. Several promising candidate biomarkers in circulating blood (miR-25-3p and miR-18b-5p), urine (miR-95, miR-21, miR-19a, and miR-19b), and prostatic secretions (miR-203) have been identified. AURKA and MYCN, novel candidate biomarkers, were found to be specifically expressed in neuroendocrine PCa. The use of BTNL2 gene mutations and inflammasomes as biomarkers in immune function-mediated, inherited PCa has also been elucidated based on NGS data. Although TBI discoveries can benefit clinical performance metrics, the translational potential and the in vivo performance of TBI outcomes need to be verified. In conclusion, TBI aids in the effective clinical management of PCa; furthermore, the fate of personalized/precision medicine mostly relies on the enhanced diagnostic, prognostic, and therapeutic potential of TBI.
Collapse
Affiliation(s)
- Vinoth-Kumar Lakshmanan
- Centre for Preclinical and Translational Medical Research (CPTMR), Central Research Facility (CRF), Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai, 600 116, Tamil Nadu, India; Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, 4184, United Arab Emirates.
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Young Do Jung
- Department of Biochemistry, Chonnam National University Medical School, 160 Baeksuh-Roh, Dong Gu, Gwangju, 61469, Republic of Korea
| |
Collapse
|
38
|
Liu Q, Lu F, Chen Z. Identification of MT1E as a novel tumor suppressor in hepatocellular carcinoma. Pathol Res Pract 2020; 216:153213. [PMID: 32956919 DOI: 10.1016/j.prp.2020.153213] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 09/06/2020] [Accepted: 09/08/2020] [Indexed: 11/26/2022]
Abstract
BACKGROUND Metallothioneins (MTs) involves in the tumorigenesis and prognosis of various cancers. The biological function and methylation status of MT1E in hepatocellular carcinoma (HCC) remain to be elucidated. METHODS We analyzed differentially expressed genes (DEGs) in tumor tissue samples and normal samples from The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) database, and identified the expression levels of MT1E in the HCC. Then, the expression levels and methylation status of MT1E in HCC tissues and cells were validated by qRT-PCR and methylation-specific PCR (MSP). Also, MTT, colony formation, transwell assays, and flow cytometry, as well as xenograft model, were used to assess the biological roles of MT1E in HCC. RESULTS Downregulated expression of MT1E was found in HCC tissues, and was notably correlated with an aberrant methylation level of the gene promoter. Moreover, our study verified that MT1E suppressed cell growth in vitro and vivo. Further study demonstrated that MT1E could induce apoptosis and suppress the metastasis of HCC cells. CONCLUSIONS Our results suggested that epigenetic silencing of MT1E due to promoter hypermethylation could play a vital role in HCC.
Collapse
Affiliation(s)
- Qichen Liu
- Departmentof General Surgery, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215000, China; Departmentof General Surgery, Binhai Country People's Hospital, Binhai, Jiangsu, 224500, China
| | - Feng Lu
- Departmentof General Surgery, Binhai Country People's Hospital, Binhai, Jiangsu, 224500, China
| | - Zhong Chen
- Departmentof General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226000, China.
| |
Collapse
|
39
|
Oncology Therapeutics Targeting the Metabolism of Amino Acids. Cells 2020; 9:cells9081904. [PMID: 32824193 PMCID: PMC7463463 DOI: 10.3390/cells9081904] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/12/2020] [Accepted: 08/13/2020] [Indexed: 12/19/2022] Open
Abstract
Amino acid metabolism promotes cancer cell proliferation and survival by supporting building block synthesis, producing reducing agents to mitigate oxidative stress, and generating immunosuppressive metabolites for immune evasion. Malignant cells rewire amino acid metabolism to maximize their access to nutrients. Amino acid transporter expression is upregulated to acquire amino acids from the extracellular environment. Under nutrient depleted conditions, macropinocytosis can be activated where proteins from the extracellular environment are engulfed and degraded into the constituent amino acids. The demand for non-essential amino acids (NEAAs) can be met through de novo synthesis pathways. Cancer cells can alter various signaling pathways to boost amino acid usage for the generation of nucleotides, reactive oxygen species (ROS) scavenging molecules, and oncometabolites. The importance of amino acid metabolism in cancer proliferation makes it a potential target for therapeutic intervention, including via small molecules and antibodies. In this review, we will delineate the targets related to amino acid metabolism and promising therapeutic approaches.
Collapse
|
40
|
Wouters J, Kalender-Atak Z, Minnoye L, Spanier KI, De Waegeneer M, Bravo González-Blas C, Mauduit D, Davie K, Hulselmans G, Najem A, Dewaele M, Pedri D, Rambow F, Makhzami S, Christiaens V, Ceyssens F, Ghanem G, Marine JC, Poovathingal S, Aerts S. Robust gene expression programs underlie recurrent cell states and phenotype switching in melanoma. Nat Cell Biol 2020; 22:986-998. [PMID: 32753671 DOI: 10.1038/s41556-020-0547-3] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 06/23/2020] [Indexed: 02/07/2023]
Abstract
Melanoma cells can switch between a melanocytic and a mesenchymal-like state. Scattered evidence indicates that additional intermediate state(s) may exist. Here, to search for such states and decipher their underlying gene regulatory network (GRN), we studied 10 melanoma cultures using single-cell RNA sequencing (RNA-seq) as well as 26 additional cultures using bulk RNA-seq. Although each culture exhibited a unique transcriptome, we identified shared GRNs that underlie the extreme melanocytic and mesenchymal states and the intermediate state. This intermediate state is corroborated by a distinct chromatin landscape and is governed by the transcription factors SOX6, NFATC2, EGR3, ELF1 and ETV4. Single-cell migration assays confirmed the intermediate migratory phenotype of this state. Using time-series sampling of single cells after knockdown of SOX10, we unravelled the sequential and recurrent arrangement of GRNs during phenotype switching. Taken together, these analyses indicate that an intermediate state exists and is driven by a distinct and stable 'mixed' GRN rather than being a symbiotic heterogeneous mix of cells.
Collapse
Affiliation(s)
- Jasper Wouters
- Center for Brain & Disease Research, VIB-KU Leuven, Leuven, Belgium.,Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Zeynep Kalender-Atak
- Center for Brain & Disease Research, VIB-KU Leuven, Leuven, Belgium.,Department of Human Genetics, KU Leuven, Leuven, Belgium.,Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Liesbeth Minnoye
- Center for Brain & Disease Research, VIB-KU Leuven, Leuven, Belgium.,Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Katina I Spanier
- Center for Brain & Disease Research, VIB-KU Leuven, Leuven, Belgium.,Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Maxime De Waegeneer
- Center for Brain & Disease Research, VIB-KU Leuven, Leuven, Belgium.,Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Carmen Bravo González-Blas
- Center for Brain & Disease Research, VIB-KU Leuven, Leuven, Belgium.,Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - David Mauduit
- Center for Brain & Disease Research, VIB-KU Leuven, Leuven, Belgium.,Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Kristofer Davie
- Center for Brain & Disease Research, VIB-KU Leuven, Leuven, Belgium.,Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Gert Hulselmans
- Center for Brain & Disease Research, VIB-KU Leuven, Leuven, Belgium.,Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Ahmad Najem
- Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Michael Dewaele
- Center for Cancer Biology, VIB-KU Leuven, Leuven, Belgium.,Department of Oncology, KU Leuven, Leuven, Belgium
| | - Dennis Pedri
- Center for Brain & Disease Research, VIB-KU Leuven, Leuven, Belgium.,Center for Cancer Biology, VIB-KU Leuven, Leuven, Belgium.,Department of Oncology, KU Leuven, Leuven, Belgium.,Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Florian Rambow
- Center for Cancer Biology, VIB-KU Leuven, Leuven, Belgium.,Department of Oncology, KU Leuven, Leuven, Belgium
| | - Samira Makhzami
- Center for Brain & Disease Research, VIB-KU Leuven, Leuven, Belgium.,Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Valerie Christiaens
- Center for Brain & Disease Research, VIB-KU Leuven, Leuven, Belgium.,Department of Human Genetics, KU Leuven, Leuven, Belgium
| | | | - Ghanem Ghanem
- Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Jean-Christophe Marine
- Center for Cancer Biology, VIB-KU Leuven, Leuven, Belgium.,Department of Oncology, KU Leuven, Leuven, Belgium
| | | | - Stein Aerts
- Center for Brain & Disease Research, VIB-KU Leuven, Leuven, Belgium. .,Department of Human Genetics, KU Leuven, Leuven, Belgium.
| |
Collapse
|
41
|
Network of clinically-relevant lncRNAs-mRNAs associated with prognosis of hepatocellular carcinoma patients. Sci Rep 2020; 10:11124. [PMID: 32636408 PMCID: PMC7341759 DOI: 10.1038/s41598-020-67742-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 06/12/2020] [Indexed: 12/16/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are often aberrantly expressed in Hepatocellular Carcinoma (HCC). We hypothesize that lncRNAs modulate HCC prognoses through differential deregulation of key lncRNAs affecting important gene network in key cancer pathways associated with pertinent clinical phenotype. Here, we present a novel approach integrating lncRNA-mRNA expression profiles with clinical characteristics to identify lncRNA signatures in clinically-relevant co-expression lncRNA-mRNA networks residing in pertinent cancer pathways. Notably one network, associated with poorer prognosis, comprises five up-regulated lncRNAs significantly correlated (|Pearson Correlation Coefficient|≥ 0.9) with 91 up-regulated genes in the cell-cycle and Rho-GTPase pathways. All 5 lncRNAs and 85/91 (93.4%) of the correlated genes were significantly associated with higher tumor-grade while 3/5 lncRNAs were also associated with no tumor capsule. Interestingly, 2/5 lncRNAs that are correlated with numerous genes in this oncogenic network were experimentally shown to up-regulate genes involved in cell-cycle and transcriptional regulation. Another network comprising 4 down-regulated lncRNAs and 8 down-regulated metallothionein-family genes are significantly associated with tumor invasion. The identification of these key lncRNAs signatures that deregulate important network of genes in key cancer pathways associated with pertinent clinical phenotype may facilitate the design of novel therapeutic strategies targeting these 'master' regulators for better patient outcome.
Collapse
|
42
|
Agioutantis PC, Loutrari H, Kolisis FN. Computational Analysis of Transcriptomic and Proteomic Data for Deciphering Molecular Heterogeneity and Drug Responsiveness in Model Human Hepatocellular Carcinoma Cell Lines. Genes (Basel) 2020; 11:E623. [PMID: 32517019 PMCID: PMC7349788 DOI: 10.3390/genes11060623] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/21/2020] [Accepted: 06/02/2020] [Indexed: 12/11/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is associated with high mortality due to its inherent heterogeneity, aggressiveness, and limited therapeutic regimes. Herein, we analyzed 21 human HCC cell lines (HCC lines) to explore intertumor molecular diversity and pertinent drug sensitivity. We used an integrative computational approach based on exploratory and single-sample gene-set enrichment analysis of transcriptome and proteome data from the Cancer Cell Line Encyclopedia, followed by correlation analysis of drug-screening data from the Cancer Therapeutics Response Portal with curated gene-set enrichment scores. Acquired results classified HCC lines into two groups, a poorly and a well-differentiated group, displaying lower/higher enrichment scores in a "Specifically Upregulated in Liver" gene-set, respectively. Hierarchical clustering based on a published epithelial-mesenchymal transition gene expression signature further supported this stratification. Between-group comparisons of gene and protein expression unveiled distinctive patterns, whereas downstream functional analysis significantly associated differentially expressed genes with crucial cancer-related biological processes/pathways and revealed concrete driver-gene signatures. Finally, correlation analysis highlighted a diverse effectiveness of specific drugs against poorly compared to well-differentiated HCC lines, possibly applicable in clinical research with patients with analogous characteristics. Overall, this study expanded the knowledge on the molecular profiles, differentiation status, and drug responsiveness of HCC lines, and proposes a cost-effective computational approach to precision anti-HCC therapies.
Collapse
Affiliation(s)
- Panagiotis C. Agioutantis
- Biotechnology Laboratory, School of Chemical Engineering, National Technical University of Athens, 5 Iroon Polytechniou Str., Zografou Campus, 15780 Athens, Greece;
- G.P. Livanos and M. Simou Laboratories, 1st Department of Critical Care Medicine & Pulmonary Services, Evangelismos Hospital, Medical School, National Kapodistrian University of Athens, 3 Ploutarchou Str., 10675 Athens, Greece
| | - Heleni Loutrari
- G.P. Livanos and M. Simou Laboratories, 1st Department of Critical Care Medicine & Pulmonary Services, Evangelismos Hospital, Medical School, National Kapodistrian University of Athens, 3 Ploutarchou Str., 10675 Athens, Greece
| | - Fragiskos N. Kolisis
- Biotechnology Laboratory, School of Chemical Engineering, National Technical University of Athens, 5 Iroon Polytechniou Str., Zografou Campus, 15780 Athens, Greece;
| |
Collapse
|
43
|
Zyxin (ZYX) promotes invasion and acts as a biomarker for aggressive phenotypes of human glioblastoma multiforme. J Transl Med 2020; 100:812-823. [PMID: 31949244 DOI: 10.1038/s41374-019-0368-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 11/21/2019] [Accepted: 12/03/2019] [Indexed: 12/17/2022] Open
Abstract
Glioblastoma multiforme (GBM) is characterized by highly invasive growth, which leads to extensive infiltration and makes complete tumor excision difficult. Since cytoskeleton proteins are related to leading processes and cell motility, and through analysis of public GBM databases, we determined that an actin-interacting protein, zyxin (ZYX), may involved in GBM invasion. Our own glioma cohort as well as the cancer genome atlas (TCGA), Rembrandt, and Gravendeel databases consistently showed that increased ZYX expression was related to tumor progression and poor prognosis of glioma patients. In vitro and in vivo experiments further confirmed the oncogenic roles of ZYX and demonstrated the role of ZYX in GBM invasive growth. Moreover, RNA-seq and mass-spectrum data from GBM cells with or without ZYX revealed that stathmin 1 (STMN1) was a potential target of ZYX. Subsequently, we found that both mRNA and protein levels of STMN1 were positively regulated by ZYX. Functionally, STMN1 not only promoted invasion of GBM cells but also rescued the invasion repression caused by ZYX loss. Taken together, our results indicate that high ZYX expression was associated with worse prognosis and highlighted that the ZYX-STMN1 axis might be a potential therapeutic target for GBM.
Collapse
|
44
|
Panchy N, Azeredo-Tseng C, Luo M, Randall N, Hong T. Integrative Transcriptomic Analysis Reveals a Multiphasic Epithelial-Mesenchymal Spectrum in Cancer and Non-tumorigenic Cells. Front Oncol 2020; 9:1479. [PMID: 32038999 PMCID: PMC6987415 DOI: 10.3389/fonc.2019.01479] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 12/09/2019] [Indexed: 12/12/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT), the conversion between rigid epithelial cells and motile mesenchymal cells, is a reversible cellular process involved in tumorigenesis, metastasis, and chemoresistance. Numerous studies have found that several types of tumor cells show a high degree of cell-to-cell heterogeneity in terms of their gene expression signatures and cellular phenotypes related to EMT. Recently, the prevalence and importance of partial or intermediate EMT states have been reported. It is unclear, however, whether there is a general pattern of cancer cell distribution in terms of the overall expression of epithelial-related genes and mesenchymal-related genes, and how this distribution is related to EMT process in normal cells. In this study, we performed integrative transcriptomic analysis that combines cancer cell transcriptomes, time course data of EMT in non-tumorigenic epithelial cells, and epithelial cells with perturbations of key EMT factors. Our statistical analysis shows that cancer cells are widely distributed in the EMT spectrum, and the majority of these cells can be described by an EMT path that connects the epithelial and the mesenchymal states via a hybrid expression region in which both epithelial genes and mesenchymal genes are highly expressed overall. We found that key patterns of this EMT path are observed in EMT progression in non-tumorigenic cells and that transcription factor ZEB1 plays a key role in defining this EMT path via diverse gene regulatory circuits connecting to epithelial genes. We performed Gene Set Variation Analysis to show that the cancer cells at hybrid EMT states also possess hybrid cellular phenotypes with both high migratory and high proliferative potentials. Our results reveal critical patterns of cancer cells in the EMT spectrum and their relationship to the EMT process in normal cells, and provide insights into the mechanistic basis of cancer cell heterogeneity and plasticity.
Collapse
Affiliation(s)
- Nicholas Panchy
- Department of Biochemistry & Cellular and Molecular Biology, The University of Tennessee, Knoxville, Knoxville, TN, United States
- National Institute for Mathematical and Biological Synthesis, Knoxville, TN, United States
| | - Cassandra Azeredo-Tseng
- Department of Biochemistry, New College of Florida, Sarasota, FL, United States
- Department of Applied Mathematics, New College of Florida, Sarasota, FL, United States
| | - Michael Luo
- Department of Mathematics & Statistics, The College of New Jersey, Ewing Township, NJ, United States
| | - Natalie Randall
- Department of Mathematics and Computer Science, Austin College, Sherman, TX, United States
| | - Tian Hong
- Department of Biochemistry & Cellular and Molecular Biology, The University of Tennessee, Knoxville, Knoxville, TN, United States
- National Institute for Mathematical and Biological Synthesis, Knoxville, TN, United States
| |
Collapse
|
45
|
Gritsenko PG, Atlasy N, Dieteren CEJ, Navis AC, Venhuizen JH, Veelken C, Schubert D, Acker-Palmer A, Westerman BA, Wurdinger T, Leenders W, Wesseling P, Stunnenberg HG, Friedl P. p120-catenin-dependent collective brain infiltration by glioma cell networks. Nat Cell Biol 2020; 22:97-107. [PMID: 31907411 PMCID: PMC6952556 DOI: 10.1038/s41556-019-0443-x] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 11/26/2019] [Indexed: 12/23/2022]
Abstract
Diffuse brain infiltration by glioma cells causes detrimental disease progression, but its multicellular coordination is poorly understood. We show here that glioma cells infiltrate the brain collectively as multicellular networks. Contacts between moving glioma cells are adaptive epithelial-like or filamentous junctions stabilized by N-cadherin, β-catenin and p120-catenin, which undergo kinetic turnover, transmit intercellular calcium transients and mediate directional persistence. Downregulation of p120-catenin compromises cell-cell interaction and communication, disrupts collective networks, and both the cadherin and RhoA binding domains of p120-catenin are required for network formation and migration. Deregulating p120-catenin further prevents diffuse glioma cell infiltration of the mouse brain with marginalized microlesions as the outcome. Transcriptomics analysis has identified p120-catenin as an upstream regulator of neurogenesis and cell cycle pathways and a predictor of poor clinical outcome in glioma patients. Collective glioma networks infiltrating the brain thus depend on adherens junctions dynamics, the targeting of which may offer an unanticipated strategy to halt glioma progression.
Collapse
Affiliation(s)
- Pavlo G Gritsenko
- Department of Cell Biology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nader Atlasy
- Department of Molecular Biology, Radboud University, Nijmegen, The Netherlands
- Center for Molecular Medicine, University Medical Center, Utrecht, The Netherlands
| | - Cindy E J Dieteren
- Department of Cell Biology, Radboud University Medical Center, Nijmegen, The Netherlands
- Protinhi Therapeutics, Nijmegen, The Netherlands
| | - Anna C Navis
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Jan-Hendrik Venhuizen
- Department of Cell Biology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Cornelia Veelken
- Department of Cell Biology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Dirk Schubert
- Cognitive Neuroscience Department, Donders Institute, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Amparo Acker-Palmer
- Institute of Cell Biology and Neuroscience and BMLS, Goethe University Frankfurt, Frankfurt, Germany
- Max Planck Institute for Brain Research, Frankfurt, Germany
| | - Bart A Westerman
- Department of Neurosurgery, VU University Medical Center, Amsterdam, The Netherlands
| | - Thomas Wurdinger
- Department of Neurosurgery, VU University Medical Center, Amsterdam, The Netherlands
| | - William Leenders
- Department of Biochemistry, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Pieter Wesseling
- Department of Pathology, Amsterdam University Medical Centers/VUmc and Brain Tumor Center Amsterdam, Amsterdam, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Hendrik G Stunnenberg
- Department of Molecular Biology, Radboud University, Nijmegen, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Peter Friedl
- Department of Cell Biology, Radboud University Medical Center, Nijmegen, The Netherlands.
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Cancer Genomics Center, Utrecht, The Netherlands.
| |
Collapse
|
46
|
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: 4.0] [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
|
47
|
Luo Y, Zeng G, Wu S. Identification of Microenvironment-Related Prognostic Genes in Bladder Cancer Based on Gene Expression Profile. Front Genet 2019; 10:1187. [PMID: 31824575 PMCID: PMC6883806 DOI: 10.3389/fgene.2019.01187] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 10/25/2019] [Indexed: 12/31/2022] Open
Abstract
Background and Objective: Bladder cancer is the most common tumor in the urinary system, with a higher incidence in men than in women and a high recurrence rate. However, the mechanism of recurrence is still unclear, and it is urgent to clarify the pathophysiological mechanism of bladder cancer. To provide theoretical basis for the development of new therapies, investigating the effect of tumor microenvironment on the prognosis of bladder cancer is necessary. Methods: We applied the Estimation of STromal and Immune cells in MAlignant Tumors using Expression data (ESTIMATE) algorithm to the downloaded TCGA (The Cancer Genome Atlas) transcriptome data to obtain the immune scores and stromal scores of each sample, and then divided the samples into two groups: high and low immune scores (or high and low stromal scores), and found that some differential genes were associated with poor prognosis of patients. We then performed protein-protein interaction (PPI) network analysis to explore the relationship between these differentially expressed genes. Moreover, we also performed (Gene Ontology) GO and (Kyoto Encyclopedia of Genes and Genomes) KEGG analyses to explore the potential functions of differentially expressed genes. Finally, our results were validated in an independent dataset. Results: We identified 136 tumor microenvironment-related genes associated with poor prognosis of bladder cancer. GO annotation and KEGG pathway enrichment analysis found that these genes are mainly involved in extracellular matrix, Focal adhesion and phosphatidylinositol 3 kinase-protein kinaseB (PI3k-Akt) signaling pathway. Next, PPI network analysis revealed some hub genes including Versican (VCAN), Thrombospondin 1 (THBS1) and Thrombospondin 1 (THBS2). Finally, 27 genes were further verified in the independent data set. Conclusions: We found 27 tumor microenvironment-related genes of bladder cancer, which are associated with poor prognosis of bladder cancer. These genes may inspire researchers to develop new treatments for bladder cancer.
Collapse
Affiliation(s)
- Yongxiang Luo
- Department of Urological Surgery, The Affiliated Luohu Hospital of Anhui University of Science and Technology, Shenzhen, China
| | - Guohua Zeng
- Department of Urological Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Song Wu
- Department of Urological Surgery, The Affiliated Luohu Hospital of Anhui University of Science and Technology, Shenzhen, China.,Shenzhen Following Precision Medical Institute, Shenzhen Luohu Hospital Group, Shenzhen, China
| |
Collapse
|
48
|
Cui Y, Zhang L, Wang W, Ma S, Liu H, Zang X, Zhang Y, Guan F. Downregulation of nicotinamide N-methyltransferase inhibits migration and epithelial-mesenchymal transition of esophageal squamous cell carcinoma via Wnt/β-catenin pathway. Mol Cell Biochem 2019; 460:93-103. [PMID: 31278587 DOI: 10.1007/s11010-019-03573-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 06/21/2019] [Indexed: 01/27/2023]
Abstract
Nicotinamide N-methyltransferase (NNMT) is an important methyltransferase involved in the biotransformation of many drugs and exogenous compounds. Abnormal expression of NNMT protein is closely associated with the onset and progression of many malignancies, but little is known about its role in esophageal squamous cell carcinoma (ESCC). Therefore, we aimed to explore whether NNMT plays any roles in carcinogenesis and metastasis in ESCC. NNMT expression was determined by immunohistochemistry in ESCC and corresponding adjacent normal tissues. Functional experiments were performed to elucidate the effects of NNMT knockdown on the proliferation, apoptosis, cell cycle, migration, and epithelial-mesenchymal transition (EMT) in EC9706 and TE1 cells. NNMT expression was significantly elevated in ESCC tissues compared with corresponding adjacent normal tissues. Moreover, a significant association emerged between NNMT expression and lymph node metastasis. SiRNA-mediated knockdown of NNMT in ESCC cells can significantly suppress cell viability and migration, induce cell cycle arrest, and promote cell apoptosis. In addition, NNMT downregulation led to the reversal of EMT, as reflected by upregulation of the intercellular adhesion molecule E-cadherin and downregulation of the mesenchymal markers N-cadherin and Vimentin. Further study found that NNMT knockdown suppressed the Wnt/β-catenin signaling pathway. Taken together, these findings indicate that NNMT is a critical regulator of EMT in ESCC and may be a potential therapeutic target for ESCC metastasis.
Collapse
Affiliation(s)
- Yanyan Cui
- College of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Luyu Zhang
- College of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Wenjie Wang
- College of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Shanshan Ma
- College of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Hongtao Liu
- College of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Xingxing Zang
- Department of Microbiology and Immunology, Einstein College of Medicine, 1300 Morris Park Ave, Bronx, New York, NY, 10461, USA
| | - Yanting Zhang
- College of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| | - Fangxia Guan
- College of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- Henan Provincial People's Hospital, Zhengzhou, 450003, China.
| |
Collapse
|
49
|
Hah YS, Cho HY, Jo SY, Park YS, Heo EP, Yoon TJ. Nicotinamide N‑methyltransferase induces the proliferation and invasion of squamous cell carcinoma cells. Oncol Rep 2019; 42:1805-1814. [PMID: 31545452 PMCID: PMC6787961 DOI: 10.3892/or.2019.7315] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 08/14/2019] [Indexed: 12/21/2022] Open
Abstract
Cutaneous squamous cell carcinoma (cSCC) is a common malignancy initiated by keratinocytes of the epidermis, which are able to invade the dermis and its periphery. Although most patients with cSCC present with curable localized tumors, recurrence, metastasis and mortality occasionally occur. In the present study, nicotinamide N‑methyltransferase (NNMT) was identified as an upregulated protein in the SCC12 cell line, which has high invasive potential compared with the SCC13 cell line. The effects of NNMT knockdown on proliferation, migration and invasion were investigated using SCC cells. shRNA‑mediated downregulation of NNMT expression levels inhibited the proliferation and density‑dependent growth of SCC12 cells. In addition, the results of a cell motility assay showed that the migration and invasion of SCC cells were markedly decreased in NNMT‑knockdown cells. The assessment of epithelial‑mesenchymal transition (EMT)‑associated gene expression using PCR array analysis revealed that high NNMT expression levels were accompanied by high expression levels of EMT‑associated genes, and that NNMT knockdown effectively suppressed the expression of matrix metalloproteinase 9, osteopontin, versican core protein and zinc finger protein SNAI2 in SCC12 cells. These results revealed that the upregulation of NNMT induced cellular invasion via EMT‑related gene expression in SCC cells.
Collapse
Affiliation(s)
- Young-Sool Hah
- Biomedical Research Institute, Gyeongsang National University Hospital, and Institute of Health Sciences, Jinju 52727, Republic of Korea
| | - Hee Young Cho
- Biomedical Research Institute, Gyeongsang National University Hospital, and Institute of Health Sciences, Jinju 52727, Republic of Korea
| | - Sun Young Jo
- Department of Dermatology, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon 51353, Republic of Korea
| | - Young Sook Park
- Department of Physical Medicine and Rehabilitation, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon 51353, Republic of Korea
| | - Eun Phil Heo
- Department of Dermatology, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon 51353, Republic of Korea
| | - Tae-Jin Yoon
- Department of Dermatology and Institute of Health Sciences, School of Medicine, Gyeongsang National University and Hospital, Jinju 52727, Republic of Korea
| |
Collapse
|
50
|
Li J, You S, Zhang S, Hu Q, Wang F, Chi X, Zhao W, Xie C, Zhang C, Yu Y, Liu J, Zhao Y, Liu P, Zhang Y, Wei X, Li Q, Wang X, Yin Z. Elevated N-methyltransferase expression induced by hepatic stellate cells contributes to the metastasis of hepatocellular carcinoma via regulation of the CD44v3 isoform. Mol Oncol 2019; 13:1993-2009. [PMID: 31294922 PMCID: PMC6717763 DOI: 10.1002/1878-0261.12544] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 06/17/2019] [Accepted: 07/10/2019] [Indexed: 12/31/2022] Open
Abstract
The cross‐talk between hepatic stellate cells (HSCs) and hepatic carcinoma cells contributes to hepatocellular carcinoma (HCC) progression, but the underlying mechanism is largely unknown. We report here that activated HSCs induce upregulation of nicotinamide N‐methyltransferase (NNMT), which is known to regulate multiple metabolic pathways in hepatoma cells of the liver. High levels of NNMT in HCC tissues were positively correlated with vascular invasion, increased serum HBV‐DNA levels, and distant metastasis. In addition, functional assays showed that NNMT promoted HCC cell invasion and metastasis by altering the histone H3 methylation on 27 methylation pattern and transcriptionally activating cluster of differentiation 44 (CD44). NNMT‐mediated N6‐methyladenosine modification of CD44 mRNA resulted in the formation of a CD44v3 splice variant, while its product 1‐methyl‐nicotinamide stabilized CD44 protein by preventing ubiquitin‐mediated degradation. Finally, NNMT was also shown to be a target of statins that inhibited metastasis of hepatoma cells. Taken together, our study shows for the first time that the NNMT/CD44v3 axis regulates HCC metastasis and presents NNMT as a promising prognostic biomarker and therapeutic target for HCC.
Collapse
Affiliation(s)
- Jie Li
- Department of Hepatobiliary Surgery, ZhongShan Hospital of Xiamen University, Fujian, China.,Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, ZhongShan Hospital of Xiamen University, Fujian, China
| | - Song You
- Graduate College of Fujian Medical University, Fuzhou, Fujian, China
| | - Sheng Zhang
- Department of Pathology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qing Hu
- Medicine Clinical Laboratory, Xiamen Xianyue Hospital, Fujian, China
| | - Fuqiang Wang
- Department of Hepatobiliary Surgery, ZhongShan Hospital of Xiamen University, Fujian, China.,Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, ZhongShan Hospital of Xiamen University, Fujian, China
| | - Xiaoqin Chi
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, ZhongShan Hospital of Xiamen University, Fujian, China
| | - Wenxiu Zhao
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, ZhongShan Hospital of Xiamen University, Fujian, China
| | - Chengrong Xie
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, ZhongShan Hospital of Xiamen University, Fujian, China
| | - Changmao Zhang
- Graduate College of Fujian Medical University, Fuzhou, Fujian, China
| | - Yaqi Yu
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, ZhongShan Hospital of Xiamen University, Fujian, China
| | - Jianmin Liu
- Department of Hepatobiliary Surgery, ZhongShan Hospital of Xiamen University, Fujian, China.,Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, ZhongShan Hospital of Xiamen University, Fujian, China
| | - Yue Zhao
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, ZhongShan Hospital of Xiamen University, Fujian, China
| | - Pingguo Liu
- Department of Hepatobiliary Surgery, ZhongShan Hospital of Xiamen University, Fujian, China.,Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, ZhongShan Hospital of Xiamen University, Fujian, China
| | - Yi Zhang
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, ZhongShan Hospital of Xiamen University, Fujian, China
| | - Xujin Wei
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, ZhongShan Hospital of Xiamen University, Fujian, China
| | - Qiu Li
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, ZhongShan Hospital of Xiamen University, Fujian, China
| | - Xiaomin Wang
- Department of Hepatobiliary Surgery, ZhongShan Hospital of Xiamen University, Fujian, China.,Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, ZhongShan Hospital of Xiamen University, Fujian, China
| | - Zhenyu Yin
- Department of Hepatobiliary Surgery, ZhongShan Hospital of Xiamen University, Fujian, China.,Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, ZhongShan Hospital of Xiamen University, Fujian, China
| |
Collapse
|