1
|
Chiang YF, Huang KC, Huang TC, Chen HY, Ali M, Al-Hendy A, Huang PS, Hsia SM. Regulatory roles of NAMPT and NAD + metabolism in uterine leiomyoma progression: Implications for ECM accumulation, stemness, and microenvironment. Redox Biol 2024; 78:103411. [PMID: 39486360 DOI: 10.1016/j.redox.2024.103411] [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: 08/30/2024] [Revised: 10/04/2024] [Accepted: 10/22/2024] [Indexed: 11/04/2024] Open
Abstract
Uterine leiomyoma (UL), commonly referred to as benign tumors, is characterized by excessive cell proliferation, extracellular matrix (ECM) accumulation, and the presence of stem cell-like properties. Nicotinamide adenine dinucleotide (NAD+) metabolism, regulated in part by nicotinamide phosphoribosyltransferase (NAMPT), plays a crucial role in these pathological processes and has emerged as a potential therapeutic target. Additionally, redox signaling pathways are integral to the pathogenesis of UL, influencing the dynamics of NAD+ metabolism. This study sought to elucidate the regulatory functions of NAMPT and NAD+ metabolism, in conjunction with redox signaling, in the progression of UL, and to explore potential therapeutic strategies targeting these pathways. Evaluation of NAMPT expression in human UL tissues revealed a positive correlation between elevated NAMPT levels and increased ECM deposition, as well as the expression of stemness markers. The use of FK866 and nicotinamide (NAM), to inhibit NAMPT significantly suppressed UL cell viability and attenuated stem cell-like characteristics. Redox signaling pathways, including those associated with DNA damage, lysosomal function homeostasis, and redox-sensitive phagophore formation, were implicated in the regulation of ECM dynamics, particularly through ECM-targeted inhibition. This study highlights the pivotal roles of NAMPT, NAD+ metabolism, and redox signaling in the pathophysiology of UL. Targeting NAMPT, particularly through the use of inhibitors FK866 and NAM, represents a promising therapeutic approach for mitigating UL progression by modulating redox and ECM dynamics. These findings offer novel insights into UL pathogenesis and establish NAMPT as a compelling target for future clinical investigation.
Collapse
Affiliation(s)
- Yi-Fen Chiang
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei, 11031, Taiwan
| | - Ko-Chieh Huang
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei, 11031, Taiwan
| | - Tsui-Chin Huang
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan
| | - Hsin-Yuan Chen
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan
| | - Mohamed Ali
- Clinical Pharmacy Department, Faculty of Pharmacy, Ain Shams University, 11566, Cairo, Egypt; Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL, 60637, USA
| | - Ayman Al-Hendy
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL, 60637, USA
| | - Pei-Shen Huang
- Department of Obstetrics and Gynecology, Taipei Medical University Hospital, Taipei, 11031, Taiwan
| | - Shih-Min Hsia
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei, 11031, Taiwan; Graduate Institute of Metabolism and Obesity Sciences, College of Nutrition, Taipei Medical University, Taipei, 11031, Taiwan; School of Food Safety, College of Nutrition, Taipei Medical University, Taipei, 11031, Taiwan; Nutrition Research Center, Taipei Medical University Hospital, Taipei, 11031, Taiwan; TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei, 11031, Taiwan.
| |
Collapse
|
2
|
Li M, Zhang L, Pan L, Zhou P, Yu R, Zhang Z, Lv J, Guo H, Wang Y, Xiao S, Liu X. Nicotinamide Efficiently Suppresses Porcine Epidemic Diarrhea Virus and Porcine Deltacoronavirus Replication. Viruses 2023; 15:1591. [PMID: 37515276 PMCID: PMC10386100 DOI: 10.3390/v15071591] [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: 06/08/2023] [Revised: 07/17/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) and porcine deltacoronavirus (PDCoV), members of the genus Coronavirus, mainly cause acute diarrhea, vomiting and dehydration in piglets, and thus lead to serious economic losses. In this study, we investigated the effects of nicotinamide (NAM) on PEDV and PDCoV replication and found that NAM treatment significantly inhibited PEDV and PDCoV reproduction. Moreover, NAM plays an important role in replication processes. NAM primarily inhibited PEDV and PDCoV RNA and protein synthesis rather than other processes. Furthermore, we discovered that NAM treatment likely inhibits the replication of PEDV and PDCoV by downregulating the expression of transcription factors through activation of the ERK1/2/MAPK pathway. Overall, this study is the first to suggest that NAM might be not only an important antiviral factor for swine intestinal coronavirus, but also a potential candidate to be evaluated in the context of other human and animal coronaviruses.
Collapse
Affiliation(s)
- Mingxia Li
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Liping Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | - Li Pan
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | - Peng Zhou
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | - Ruiming Yu
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | - Zhongwang Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | - Jianliang Lv
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | - Huichen Guo
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | - Yonglu Wang
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | - Sa Xiao
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Xinsheng Liu
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| |
Collapse
|
3
|
Hunt SV, Jamison A, Malhotra R. Oral nicotinamide for non-melanoma skin cancers: A review. Eye (Lond) 2023; 37:823-829. [PMID: 35347291 PMCID: PMC10050186 DOI: 10.1038/s41433-022-02036-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/08/2022] [Accepted: 03/15/2022] [Indexed: 12/15/2022] Open
Abstract
There has been much interest in the role of oral nicotinamide supplementation in reducing the incidence of non-melanoma skin cancers. This article reviews the hypothesised mechanisms of action of nicotinamide, and the available literature outlining its role for this purpose. There have been five randomised controlled trials (RCT), one histopathological study and two case series exploring the effect of oral nicotinamide supplementation on UV-induced immunosuppression of the skin, and incidence of actinic keratoses and non-melanoma skin cancers (NMSC). The largest RCT received criticism of the statistical analyses used, but the critics still acknowledged a likely benefit of treatment with oral nicotinamide in reducing the incidence of NMSC. Nicotinamide has a favourable safety profile. Current evidence is not definitive that oral nicotinamide supplementation reduces the incidence of NMSC, but it constitutes a low-risk management option that may be particularly relevant for high-risk individuals, and should be discussed as an option for these patients.
Collapse
Affiliation(s)
| | - Aaron Jamison
- Corneoplastic unit, Queen Victoria Hospital, East Grinstead, England
| | - Raman Malhotra
- Corneoplastic unit, Queen Victoria Hospital, East Grinstead, England
| |
Collapse
|
4
|
Gao L, Du X, Li J, Qin FXF. Evolving roles of CD38 metabolism in solid tumour microenvironment. Br J Cancer 2023; 128:492-504. [PMID: 36396822 PMCID: PMC9938187 DOI: 10.1038/s41416-022-02052-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/20/2022] [Accepted: 10/27/2022] [Indexed: 11/19/2022] Open
Abstract
Given that plenty of clinical findings and reviews have already explained in detail on the progression of CD38 in multiple myeloma and haematological system tumours, here we no longer give unnecessary discussion on the above progression. Though therapeutic antibodies have been regarded as a greatest breakthrough in multiple myeloma immunotherapies due to the durable anti-tumour responses in the clinic, but the role of CD38 in the immunologic regulation and evasion of non-hematopoietic solid tumours are just initiated and controversial. Therefore, we will focus on the bio-function of CD38 enzymatic substrates or metabolites in the variety of non-hematopoietic malignancies and the potential therapeutic value of targeting the CD38-NAD+ or CD38-cADPR/ADPR signal axis. Though limited, we review some ongoing researches and clinical trials on therapeutic approaches in solid tumour as well.
Collapse
Affiliation(s)
- Long Gao
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, 230022, Hefei, China
| | - Xiaohong Du
- Institute of Clinical Medicine Research, Suzhou Science and Technology Town Hospital, Suzhou, China
| | - Jiabin Li
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, 230022, Hefei, China.
| | - F Xiao-Feng Qin
- Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 100005, Beijing, China.
- Suzhou Institute of Systems Medicine, 215123, Suzhou, China.
| |
Collapse
|
5
|
Silva HVR, da Silva GÁF, Zavan B, Machado RP, de Araujo-Neto JH, Ellena JA, Ionta M, Barbosa MIF, Doriguetto AC. The nicotinamide ruthenium(II) complex induces the production of reactive oxygen species (ROS), cell cycle arrest, and apoptosis in melanoma cells. Polyhedron 2023. [DOI: 10.1016/j.poly.2022.116267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
6
|
Wong W, Crane ED, Zhang H, Li J, Day TA, Green AE, Menzies KJ, Crane JD. Pgc-1α controls epidermal stem cell fate and skin repair by sustaining NAD + homeostasis during aging. Mol Metab 2022; 65:101575. [PMID: 35987498 PMCID: PMC9463389 DOI: 10.1016/j.molmet.2022.101575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/30/2022] [Accepted: 08/11/2022] [Indexed: 10/31/2022] Open
Abstract
OBJECTIVE The epidermal barrier is renewed by the activation, proliferation, and differentiation of keratinocyte stem cells after injury and aging impedes this repair process through undefined mechanisms. We previously identified a gene signature of metabolic dysfunction in aged murine epidermis, but the precise regulators of epidermal repair and age-related growth defects are not well established. Aged mouse models as well as mice with conditional epidermal loss of the metabolic regulator, peroxisome proliferator-activated receptor gamma coactivator-1 alpha (Pgc-1α) were used to explore the cellular pathways which control skin repair after injury and stress. METHODS Aged mice or those with epidermal Pgc-1α deletion (epiPgc-1α KO) and young or Pgc1afl/fl controls were subjected to wound injury, UVB exposure or the inflammatory agent TPA. In vivo and ex vivo analyses of wound closure, skin structure, cell growth and stem cell differentiation were used to understand changes in epidermal re-growth and repair resulting from aging or Pgc-1α loss. RESULTS Aging impairs epidermal re-growth during wound healing and results in lower expression of Pgc-1α. Mice with conditional deletion of epidermal Pgc-1α exhibit greater inflammation- and UVB-induced cell differentiation, reduced proliferation, and slower wound healing. epiPgc-1α KO mice also displayed reduced keratinocyte NAD+ levels, shorter telomeres, and greater poly ADP-ribosylation, resulting in enhanced stress-stimulated p53 and p21 signaling. When NAD+ was reduced by Pgc-1α loss or pharmacologic inhibition of NAD+ synthesis, there was reduced stress-induced proliferation, increased differentiation, and protection against DNA damage via enhanced epidermal shedding. Similarly, aged mice exhibit disrupted epidermal NAD+ homeostasis and enhanced p53 activation, resulting in p21 growth arrest after wounding. NAD+ precursor treatment restores epidermal growth from old skin to that of young. CONCLUSIONS Our studies identify a novel role for epidermal Pgc-1α in controlling epidermal repair via its regulation of cellular NAD+ and downstream effects on p53-driven growth arrest. We also establish that parallel mechanisms are evident in aged epidermis, showing that NAD+ signaling is an important controller of physiologic skin repair and that dysfunction of this pathway contributes to age-related wound repair defects.
Collapse
Affiliation(s)
- Wesley Wong
- Department of Biology, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA
| | - Elizabeth D Crane
- Department of Biology, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA
| | - Hui Zhang
- Department of Biology, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA
| | - Jiahe Li
- Department of Bioengineering, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA
| | - Tovah A Day
- Department of Biology, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA
| | - Alex E Green
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Canada
| | - Keir J Menzies
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Canada
| | - Justin D Crane
- Department of Biology, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA.
| |
Collapse
|
7
|
Mainville L, Smilga AS, Fortin PR. Effect of Nicotinamide in Skin Cancer and Actinic Keratoses Chemoprophylaxis, and Adverse Effects Related to Nicotinamide: A Systematic Review and Meta-Analysis. J Cutan Med Surg 2022; 26:297-308. [PMID: 35134311 PMCID: PMC9125143 DOI: 10.1177/12034754221078201] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Oral nicotinamide is recommended in individuals with a field of cancerization or with ≥1 previous cutaneous squamous cell carcinoma (cSCC). OBJECTIVE To evaluate the effect of nicotinamide in prevention of skin cancers. METHODS We conducted a systematic review and meta-analysis of randomized controlled trials to evaluate the effect of nicotinamide. We used Medline, EMBASE, CENTRAL, and Web of Science databases from their inception to October 2020 to search the following concepts: "nicotinamide"; "randomized controlled trial" (validated filters). Two independent reviewers screened titles and abstracts for intervention and study design before searching full texts for eligibility criteria. To be eligible, ≥1 outcome had to be covered. We used a standardized collection grid to complete data extraction in duplicate. The primary outcome was skin cancers (all types). Secondary outcomes were basal cell carcinomas (BCCs); cSCCs; actinic keratoses; melanomas; digestive, cutaneous, and biochemical adverse effects (AEs). Subgroup analyses were planned a priori. RESULTS We screened 4730 citations and found 29 trials (3039 patients) meeting inclusion criteria. Nicotinamide was associated with a significant reduction in skin cancers compared to control (rate ratio 0.50 (95% CI, 0.29-0.85; I 2 = 64%; 552 patients; 5 trials); moderate strength of the evidence). Heterogeneity was explained by risk of bias. Nicotinamide was associated with a significant reduction in BCCs and cSCCs, and increased risk of digestive AEs. CONCLUSION Oral nicotinamide should be considered in healthy patients or organ transplant recipients with history of skin cancer (GRADE: weak recommendation; moderate-quality evidence), in particular of BCC and cSCC.
Collapse
Affiliation(s)
| | | | - Paul R. Fortin
- Infectious and Immune Diseases, Centre de recherche du CHU de Québec – Université Laval, Quebec, Canada
- Division of Rheumatology, Department of Medicine, CHU de Québec – Université Laval, Quebec, Canada
| |
Collapse
|
8
|
Perez M, Abisaad JA, Rojas KD, Marchetti MA, Jaimes N. Skin Cancer: Primary, Secondary, and Tertiary Prevention. Part I. J Am Acad Dermatol 2022; 87:255-268. [DOI: 10.1016/j.jaad.2021.12.066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 12/03/2021] [Accepted: 12/15/2021] [Indexed: 10/19/2022]
|
9
|
Elmets CA, Slominski A, Athar M. The Challenge of Melanoma Chemoprevention. Cancer Prev Res (Phila) 2022; 15:71-74. [PMID: 35027465 PMCID: PMC9306336 DOI: 10.1158/1940-6207.capr-21-0595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/04/2022] [Accepted: 01/06/2022] [Indexed: 01/07/2023]
Abstract
Melanoma is a treatment-resistant cancer of melanocytes. There is a serious unmet need for chemopreventive agents that can inhibit their evolution from preexisting dysplastic nevi. Low-dose aspirin and NSAIDs are potential chemopreventive candidates because they inhibit the enzyme COX-2 which has a number of procarcinogenic effects. Unfortunately, the clinical trial reported by Okwundu and colleagues in this issue of Cancer Prevention Research did not show an effect of aspirin on biomarkers associated with progression of premalignant dysplastic nevi to melanomas. Further clinical trials with other aspirin or NSAID biomarkers or clinical trials with other potential chemopreventive agents offer hope to those who are at increased risk for melanomas.See related article, p. 129.
Collapse
Affiliation(s)
- Craig A. Elmets
- Department of Dermatology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama.,Department of Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama.,O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama.,Birmingham VA Medical Center, Birmingham, Alabama.,Corresponding Author: Craig A. Elmets, Department of Dermatology, University of Alabama at Birmingham, 510 20 Street South, FOT Suite 858, Birmingham, AL 35233. Phone: 205-934-5188; Fax: 205-934-5766; E-mail:
| | - Andrzej Slominski
- Department of Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama.,O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama.,Birmingham VA Medical Center, Birmingham, Alabama
| | - Mohammad Athar
- Department of Dermatology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama.,O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
10
|
Alavi S, Emran AA, Tseng HY, Tiffen JC, McGuire HM, Hersey P. Nicotinamide Inhibits T Cell Exhaustion and Increases Differentiation of CD8 Effector T Cells. Cancers (Basel) 2022; 14:cancers14020323. [PMID: 35053490 PMCID: PMC8774026 DOI: 10.3390/cancers14020323] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/06/2022] [Accepted: 01/07/2022] [Indexed: 12/11/2022] Open
Abstract
One of the limitations of immunotherapy is the development of a state referred to as T cell exhaustion (TEx) whereby T cells express inhibitory receptors (IRs) and lose production of effectors involved in killing of their targets. In the present studies we have used the repeated stimulation model with anti CD3 and anti CD28 to understand the factors involved in TEx development and treatments that may reduce changes of TEx. The results show that addition of nicotinamide (NAM) involved in energy supply to cells prevented the development of inhibitory receptors (IRs). This was particularly evident for the IRs CD39, TIM3, and to a lesser extent LAG3 and PD1 expression. NAM also prevented the inhibition of IL-2 and TNFα expression in TEx and induced differentiation of CD4+ and CD8 T cells to effector memory and terminal effector T cells. The present results showed that effects of NAM were linked to regulation of reactive oxygen species (ROS) consistent with previous studies implicating ROS in upregulation of TOX transcription factors that induce TEx. These effects of NAM in reducing changes of TEx and in increasing the differentiation of T cells to effector states appears to have important implications for the use of NAM supplements in immunotherapy against cancers and viral infections and require further exploration in vivo.
Collapse
Affiliation(s)
- Sara Alavi
- Melanoma Immunology and Oncology Program, The Centenary Institute, University of Sydney, Camperdown 2050, Australia; (S.A.); (A.A.E.); (H.-Y.T.); (J.C.T.)
- Melanoma Institute Australia, Crows Nest, Sydney 2065, Australia
- Melanoma Epigenetics Lab, The Centenary Institute, University of Sydney, Camperdown 2050, Australia
| | - Abdullah Al Emran
- Melanoma Immunology and Oncology Program, The Centenary Institute, University of Sydney, Camperdown 2050, Australia; (S.A.); (A.A.E.); (H.-Y.T.); (J.C.T.)
- Melanoma Institute Australia, Crows Nest, Sydney 2065, Australia
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02114, USA
| | - Hsin-Yi Tseng
- Melanoma Immunology and Oncology Program, The Centenary Institute, University of Sydney, Camperdown 2050, Australia; (S.A.); (A.A.E.); (H.-Y.T.); (J.C.T.)
- Melanoma Institute Australia, Crows Nest, Sydney 2065, Australia
- Melanoma Epigenetics Lab, The Centenary Institute, University of Sydney, Camperdown 2050, Australia
| | - Jessamy C. Tiffen
- Melanoma Immunology and Oncology Program, The Centenary Institute, University of Sydney, Camperdown 2050, Australia; (S.A.); (A.A.E.); (H.-Y.T.); (J.C.T.)
- Melanoma Institute Australia, Crows Nest, Sydney 2065, Australia
- Melanoma Epigenetics Lab, The Centenary Institute, University of Sydney, Camperdown 2050, Australia
| | - Helen Marie McGuire
- Melanoma Immunology and Oncology Program, The Centenary Institute, University of Sydney, Camperdown 2050, Australia; (S.A.); (A.A.E.); (H.-Y.T.); (J.C.T.)
- Ramaciotti Facility for Human Systems Biology, University of Sydney, Sydney 2050, Australia
- Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Sydney 2050, Australia
- Correspondence: (H.M.M.); (P.H.); Tel.: +61-2-9565-6001 (P.H.)
| | - Peter Hersey
- Melanoma Immunology and Oncology Program, The Centenary Institute, University of Sydney, Camperdown 2050, Australia; (S.A.); (A.A.E.); (H.-Y.T.); (J.C.T.)
- Melanoma Institute Australia, Crows Nest, Sydney 2065, Australia
- Correspondence: (H.M.M.); (P.H.); Tel.: +61-2-9565-6001 (P.H.)
| |
Collapse
|
11
|
Engineering of the thermophilic nitrile hydratase from Pseudonocardia thermophila JCM3095 for large-scale nicotinamide production based on sequence-activity relationships. Int J Biol Macromol 2021; 191:775-782. [PMID: 34592221 DOI: 10.1016/j.ijbiomac.2021.09.132] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 09/06/2021] [Accepted: 09/20/2021] [Indexed: 11/23/2022]
Abstract
The green biocatalyst nitrile hydratase (NHase) is able to bio-transform 3-cyanopyridine into nicotinamide. As the NHase reaction is exothermic, an enzyme with high activity and stability is needed for nicotinamide production. In this study, we used sequence analysis and site-directed mutagenesis to generate a mutant of thermophilic NHase from Pseudonocardia thermophila JCM3095 with substantially enhanced activity and developed a powerful process for nicotinamide bio-production. The specific activity of αF126Y/αF168Y mutant was successfully increased by 3.98-fold over that of the wild-type enzyme. The half-life of such mutant was longer than 2 h, which was comparable to its parent enzyme. The relative activity of the αF126Y/αF168Y mutant after treatment with 1 M 3-cyanopyridine and 2 M nicotinamide was 73.2% and 63.7%, respectively, showing minor loss of its original stability. Structural analysis demonstrated that hydrogen bonds at the active site and α-β subunit interface of the NHase contribute to the improved activity and the maintenance of stability. Escherichia coli transformant harboring the mutant NHase was used for nicotinamide bio-production, yielding a nicotinamide productivity of 251.1 g/(L·h), which is higher than the productivity obtained using other NHase-containing strains and transformants. The newly established variant is therefore a promising alternative for the industrial production of nicotinamides.
Collapse
|
12
|
Pellegrini M, D’Eusebio C, Ponzo V, Tonella L, Finocchiaro C, Fierro MT, Quaglino P, Bo S. Nutritional Interventions for Patients with Melanoma: From Prevention to Therapy-An Update. Nutrients 2021; 13:4018. [PMID: 34836273 PMCID: PMC8624488 DOI: 10.3390/nu13114018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/31/2021] [Accepted: 11/08/2021] [Indexed: 12/17/2022] Open
Abstract
Melanoma is an aggressive skin cancer, whose incidence rates have increased over the past few decades. Risk factors for melanoma are both intrinsic (genetic and familiar predisposition) and extrinsic (environment, including sun exposure, and lifestyle). The recent advent of targeted and immune-based therapies has revolutionized the treatment of melanoma, and research is focusing on strategies to optimize them. Obesity is an established risk factor for several cancer types, but its possible role in the etiology of melanoma is controversial. Body mass index, body surface area, and height have been related to the risk for cutaneous melanoma, although an 'obesity paradox' has been described too. Increasing evidence suggests the role of nutritional factors in the prevention and management of melanoma. Several studies have demonstrated the impact of dietary attitudes, specific foods, and nutrients both on the risk for melanoma and on the progression of the disease, via the effects on the oncological treatments. The aim of this narrative review was to summarize the main literature results regarding the preventive and therapeutic role of nutritional schemes, specific foods, and nutrients on melanoma incidence and progression.
Collapse
Affiliation(s)
- Marianna Pellegrini
- Department of Medical Sciences, Division of Endocrinology, Diabetes and Metabolism, University of Torino, 10126 Torino, Italy; (M.P.); (C.D.); (V.P.); (S.B.)
| | - Chiara D’Eusebio
- Department of Medical Sciences, Division of Endocrinology, Diabetes and Metabolism, University of Torino, 10126 Torino, Italy; (M.P.); (C.D.); (V.P.); (S.B.)
| | - Valentina Ponzo
- Department of Medical Sciences, Division of Endocrinology, Diabetes and Metabolism, University of Torino, 10126 Torino, Italy; (M.P.); (C.D.); (V.P.); (S.B.)
| | - Luca Tonella
- Department of Medical Sciences, Dermatologic Clinic, University of Torino, 10126 Torino, Italy; (L.T.); (M.T.F.)
| | - Concetta Finocchiaro
- Dietetic and Clinical Nutrition Unit, “Città della Salute e della Scienza” Hospital, 10126 Torino, Italy;
| | - Maria Teresa Fierro
- Department of Medical Sciences, Dermatologic Clinic, University of Torino, 10126 Torino, Italy; (L.T.); (M.T.F.)
| | - Pietro Quaglino
- Department of Medical Sciences, Dermatologic Clinic, University of Torino, 10126 Torino, Italy; (L.T.); (M.T.F.)
| | - Simona Bo
- Department of Medical Sciences, Division of Endocrinology, Diabetes and Metabolism, University of Torino, 10126 Torino, Italy; (M.P.); (C.D.); (V.P.); (S.B.)
| |
Collapse
|
13
|
Abstract
There has been considerable research in recent years on dietary factors and/or nutritional elements that might impact melanoma risk. A wide variety of dietary compounds have been studied, but only a selected group will be discussed in this review. Many have promising in vitro evidence supporting their potential, and some have been associated with decreased melanoma risk in epidemiologic studies; however, data from randomized controlled trials in humans are lacking. Future studies may be able to clarify the potential role of dietary components in melanoma risk reduction.
Collapse
|
14
|
Beyond Nicotinamide Metabolism: Potential Role of Nicotinamide N-Methyltransferase as a Biomarker in Skin Cancers. Cancers (Basel) 2021; 13:cancers13194943. [PMID: 34638427 PMCID: PMC8508019 DOI: 10.3390/cancers13194943] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/15/2021] [Accepted: 09/28/2021] [Indexed: 02/01/2023] Open
Abstract
Skin cancers (SC) collectively represent the most common type of malignancy in white populations. SC includes two main forms: malignant melanoma and non-melanoma skin cancer (NMSC). NMSC includes different subtypes, namely, basal cell carcinoma (BCC), squamous cell carcinoma (SCC), Merkel cell carcinoma (MCC), and keratoacanthoma (KA), together with the two pre-neoplastic conditions Bowen disease (BD) and actinic keratosis (AK). Both malignant melanoma and NMSC are showing an increasing incidence rate worldwide, thus representing an important challenge for health care systems, also because, with some exceptions, SC are generally characterized by an aggressive behavior and are often diagnosed late. Thus, identifying new biomarkers suitable for diagnosis, as well as for prognosis and targeted therapy is mandatory. Nicotinamide N-methyltransferase (NNMT) is an enzyme that is emerging as a crucial player in the progression of several malignancies, while its substrate, nicotinamide, is known to exert chemopreventive effects. Since there is increasing evidence regarding the involvement of this enzyme in the malignant behavior of SC, the current review aims to summarize the state of the art as concerns NNMT role in SC and to support future studies focused on exploring the diagnostic and prognostic potential of NNMT in skin malignancies and its suitability for targeted therapy.
Collapse
|
15
|
Abstract
NAD(H) and NADP(H) have traditionally been viewed as co-factors (or co-enzymes) involved in a myriad of oxidation-reduction reactions including the electron transport in the mitochondria. However, NAD pathway metabolites have many other important functions, including roles in signaling pathways, post-translational modifications, epigenetic changes, and regulation of RNA stability and function via NAD-capping of RNA. Non-oxidative reactions ultimately lead to the net catabolism of these nucleotides, indicating that NAD metabolism is an extremely dynamic process. In fact, recent studies have clearly demonstrated that NAD has a half-life in the order of minutes in some tissues. Several evolving concepts on the metabolism, transport, and roles of these NAD pathway metabolites in disease states such as cancer, neurodegeneration, and aging have emerged in just the last few years. In this perspective, we discuss key recent discoveries and changing concepts in NAD metabolism and biology that are reshaping the field. In addition, we will pose some open questions in NAD biology, including why NAD metabolism is so fast and dynamic in some tissues, how NAD and its precursors are transported to cells and organelles, and how NAD metabolism is integrated with inflammation and senescence. Resolving these questions will lead to significant advancements in the field.
Collapse
|
16
|
Morandi F, Horenstein AL, Malavasi F. The Key Role of NAD + in Anti-Tumor Immune Response: An Update. Front Immunol 2021; 12:658263. [PMID: 33936090 PMCID: PMC8082456 DOI: 10.3389/fimmu.2021.658263] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/25/2021] [Indexed: 12/16/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) is an important molecule that functions as a co-enzyme in numerous metabolic processes. Generated both through de novo synthesis and via salvage pathways, NAD+ is the substrate for a variety of NAD+-consuming enzymes. Among them is CD38, a cell surface ecto-enzyme widely expressed on different types of cells and endowed with the function of cADP-ribose synthases/NAD+ glycohydrolase. Surface CD38 expression is increased in different hematological and solid tumors, where it cooperates with other ecto-enzymes to produce the immunosuppressive molecule adenosine (ADO). Few studies have explored the correlation of NAD+ levels with T-cell mediated anti-tumor response in preclinical models. We therefore discuss these novel findings, examining the possible contribution of NAD+ depletion, along with ADO production, in the immunosuppressive activities of CD38 in the context of human tumors. Lastly, we discuss the use of pharmacological inhibitors of CD38 and supplementation of different NAD+ precursors to increase NAD+ levels and to boost T cell responses. Such molecules may be employed as adjuvant therapies, in combination with standard treatments, for cancer patients.
Collapse
Affiliation(s)
- Fabio Morandi
- Laboratorio Cellule Staminali Post-Natali e Terapie Cellulari, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Alberto Leonardo Horenstein
- Dipartimento Scienze Mediche, Università di Torino, Centro Ricerche Medicina Sperimentale (CeRMS) and Fondazione Ricerca Molinette Onlus, Torino, Italy
| | - Fabio Malavasi
- Dipartimento Scienze Mediche, Università di Torino, Centro Ricerche Medicina Sperimentale (CeRMS) and Fondazione Ricerca Molinette Onlus, Torino, Italy
| |
Collapse
|
17
|
Giacalone S, Spigariolo CB, Bortoluzzi P, Nazzaro G. Oral nicotinamide: The role in skin cancer chemoprevention. Dermatol Ther 2021; 34:e14892. [PMID: 33595161 DOI: 10.1111/dth.14892] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 02/13/2021] [Indexed: 12/28/2022]
Abstract
The incidence of skin cancer has gradually increased in the last years and exposition to ultraviolet radiation remains the main risk factor. We performed a comprehensive review on the role of nicotinamide (NAM) in the chemoprevention of skin cancers. NAM, a water-soluble form of vitamin B3, interferes with skin carcinogenesis as it regulates immunosuppressor genes such as p53 and sirtuins and restores intracellular level of NAD+, a co-enzyme essential for energy production. Efficacy and safety of NAM was evaluated in a Phase III double-blinded control-placebo study (ONTRAC), thus demonstrating that the incidence of actinic keratoses and non-melanoma skin cancers was lower in the nicotinamide group than in placebo group. Further studies showed the efficacy of NAM also in transplanted patients and among inhabitants living in arsenic contamination areas. Despite the quick response to NAM supplementation, its intake need to be carried on chronically as the efficacy seems to vanish rapidly.
Collapse
Affiliation(s)
- Serena Giacalone
- Dermatology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Cristina B Spigariolo
- Dermatology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Paolo Bortoluzzi
- Dermatology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Gianluca Nazzaro
- Dermatology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
18
|
Atkins MB, Curiel-Lewandrowski C, Fisher DE, Swetter SM, Tsao H, Aguirre-Ghiso JA, Soengas MS, Weeraratna AT, Flaherty KT, Herlyn M, Sosman JA, Tawbi HA, Pavlick AC, Cassidy PB, Chandra S, Chapman PB, Daud A, Eroglu Z, Ferris LK, Fox BA, Gershenwald JE, Gibney GT, Grossman D, Hanks BA, Hanniford D, Hernando E, Jeter JM, Johnson DB, Khleif SN, Kirkwood JM, Leachman SA, Mays D, Nelson KC, Sondak VK, Sullivan RJ, Merlino G. The State of Melanoma: Emergent Challenges and Opportunities. Clin Cancer Res 2021; 27:2678-2697. [PMID: 33414132 DOI: 10.1158/1078-0432.ccr-20-4092] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 11/26/2020] [Accepted: 01/04/2021] [Indexed: 12/17/2022]
Abstract
Five years ago, the Melanoma Research Foundation (MRF) conducted an assessment of the challenges and opportunities facing the melanoma research community and patients with melanoma. Since then, remarkable progress has been made on both the basic and clinical research fronts. However, the incidence, recurrence, and death rates for melanoma remain unacceptably high and significant challenges remain. Hence, the MRF Scientific Advisory Council and Breakthrough Consortium, a group that includes clinicians and scientists, reconvened to facilitate intensive discussions on thematic areas essential to melanoma researchers and patients alike, prevention, detection, diagnosis, metastatic dormancy and progression, response and resistance to targeted and immune-based therapy, and the clinical consequences of COVID-19 for patients with melanoma and providers. These extensive discussions helped to crystalize our understanding of the challenges and opportunities facing the broader melanoma community today. In this report, we discuss the progress made since the last MRF assessment, comment on what remains to be overcome, and offer recommendations for the best path forward.
Collapse
Affiliation(s)
- Michael B Atkins
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, D.C.
| | - Clara Curiel-Lewandrowski
- Department of Dermatology, The University of Arizona Cancer Center Skin Cancer Institute, College of Medicine, University of Arizona, Tucson, Arizona
| | - David E Fisher
- Department of Dermatology & Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Susan M Swetter
- Department of Dermatology, Pigmented Lesion & Melanoma Program, Stanford University Medical Center & Cancer Institute, VA Palo Alto Health Care System, Palo Alto, California
| | - Hensin Tsao
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Julio A Aguirre-Ghiso
- Division of Hematology & Oncology, Departments of Medicine, Otolaryngology, & Oncological Sciences, Precision Immunology Institute, Black Family Stem Cell Institute, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Maria S Soengas
- Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Ashani T Weeraratna
- Department of Biochemistry & Molecular Biology, Johns Hopkins Bloomberg School of Public Health & Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Keith T Flaherty
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Jeffrey A Sosman
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - Hussein A Tawbi
- Division of Cancer Medicine, Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Pamela B Cassidy
- Knight Cancer Institute & Department of Dermatology, Oregon Health & Science University, Portland, Oregon
| | - Sunandana Chandra
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - Paul B Chapman
- Memorial Sloan Kettering Cancer Center & Weill Cornell Medical College, New York, New York
| | - Adil Daud
- University of California, San Francisco, California
| | - Zeynep Eroglu
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Laura K Ferris
- Department of Dermatology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Bernard A Fox
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Laboratory of Molecular & Tumor Immunology, Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Cancer Institute, Portland, Oregon
| | - Jeffrey E Gershenwald
- Departments of Surgical Oncology & Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Geoffrey T Gibney
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, D.C
| | - Douglas Grossman
- Huntsman Cancer Institute & Department of Dermatology, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Brent A Hanks
- Division of Medical Oncology, Department of Medicine, Department of Pharmacology & Cancer Biology, Center for Cancer Immunotherapy, Duke University Medical Center, Durham, North Carolina
| | - Douglas Hanniford
- Department of Pathology, NYU Grossman School of Medicine, Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York, New York
| | - Eva Hernando
- Department of Pathology, NYU Grossman School of Medicine, Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York, New York
| | - Joanne M Jeter
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Douglas B Johnson
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Samir N Khleif
- The Loop Laboratory for Immuno-Oncology Lombardi Cancer Center, Georgetown School of Medicine, Georgetown University, Washington, D.C
| | | | - Sancy A Leachman
- Knight Cancer Institute & Department of Dermatology, Oregon Health & Science University, Portland, Oregon
| | - Darren Mays
- Department of Internal Medicine, College of Medicine, The Ohio State University, Center for Tobacco Research, The Ohio State University James Comprehensive Cancer Center, Columbus, Ohio
| | - Kelly C Nelson
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Vernon K Sondak
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Ryan J Sullivan
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | | | | |
Collapse
|
19
|
Selvanesan BC, Meena K, Beck A, Meheus L, Lara O, Rooman I, Gravekamp C. Nicotinamide combined with gemcitabine is an immunomodulatory therapy that restrains pancreatic cancer in mice. J Immunother Cancer 2020; 8:e001250. [PMID: 33154149 PMCID: PMC7646363 DOI: 10.1136/jitc-2020-001250] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2020] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Treatments for pancreatic ductal adenocarcinoma are poorly effective, at least partly due to the tumor's immune-suppressive stromal compartment. New evidence of positive effects on immune responses in the tumor microenvironment (TME), compelled us to test the combination of gemcitabine (GEM), a standard chemotherapeutic for pancreatic cancer, with nicotinamide (NAM), the amide form of niacin (vitamin B3), in mice with pancreatic cancer. METHODS Various mouse tumor models of pancreatic cancer, that is, orthotopic Panc-02 and KPC (KrasG12D, p53R172H, Pdx1-Cre) grafts, were treated alternately with NAM and GEM for 2 weeks, and the effects on efficacy, survival, stromal architecture and tumor-infiltrating immune cells was examined by immunohistochemistry (IHC), flow cytometry, Enzyme-linked immunospot (ELISPOT), T cell depletions in vivo, Nanostring analysis and RNAscope. RESULTS A significant reduction in tumor weight and number of metastases was found, as well as a significant improved survival of the NAM+GEM group compared with all control groups. IHC and flow cytometry showed a significant decrease in tumor-associated macrophages and myeloid-derived suppressor cells in the tumors of NAM+GEM-treated mice. This correlated with a significant increase in the number of CD4 and CD8 T cells of NAM+GEM-treated tumors, and CD4 and CD8 T cell responses to tumor-associated antigen survivin, most likely through epitope spreading. In vivo depletions of T cells demonstrated the involvement of CD4 T cells in the eradication of the tumor by NAM+GEM treatment. In addition, remodeling of the tumor stroma was observed with decreased collagen I and lower expression of hyaluronic acid binding protein, reorganization of the immune cells into lymph node like structures and CD31 positive vessels. Expression profiling for a panel of immuno-oncology genes revealed significant changes in genes involved in migration and activation of T cells, attraction of dendritic cells and epitope spreading. CONCLUSION This study highlights the potential of NAM+GEM as immunotherapy for advanced pancreatic cancer.
Collapse
Affiliation(s)
| | - Kiran Meena
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Amanda Beck
- Michael F. Price Center, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Lydie Meheus
- AntiCancer Fund, Boechoutlaan, Strombeek-Bever, Belgium
| | - Olaya Lara
- Laboratory of Medical and Molecular Oncology, Vrije Universiteit Brussel, Laarbeeklaan, Brussels, Belgium
| | - Ilse Rooman
- AntiCancer Fund, Boechoutlaan, Strombeek-Bever, Belgium
- Laboratory of Medical and Molecular Oncology, Vrije Universiteit Brussel, Laarbeeklaan, Brussels, Belgium
| | - Claudia Gravekamp
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
20
|
Nicotinamide inhibits melanoma in vitro and in vivo. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:211. [PMID: 33028392 PMCID: PMC7542872 DOI: 10.1186/s13046-020-01719-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 09/24/2020] [Indexed: 02/07/2023]
Abstract
Background Even though new therapies are available against melanoma, novel approaches are needed to overcome resistance and high-toxicity issues. In the present study the anti-melanoma activity of Nicotinamide (NAM), the amide form of Niacin, was assessed in vitro and in vivo. Methods Human (A375, SK-MEL-28) and mouse (B16-F10) melanoma cell lines were used for in vitro investigations. Viability, cell-death, cell-cycle distribution, apoptosis, Nicotinamide Adenine Dinucleotide+ (NAD+), Adenosine Triphosphate (ATP), and Reactive Oxygen Species (ROS) levels were measured after NAM treatment. NAM anti-SIRT2 activity was tested in vitro; SIRT2 expression level was investigated by in silico transcriptomic analyses. Melanoma growth in vivo was measured in thirty-five C57BL/6 mice injected subcutaneously with B16-F10 melanoma cells and treated intraperitoneally with NAM. Interferon (IFN)-γ-secreting murine cells were counted with ELISPOT assay. Cytokine/chemokine plasmatic levels were measured by xMAP technology. Niacin receptors expression in human melanoma samples was also investigated by in silico transcriptomic analyses. Results NAM reduced up to 90% melanoma cell number and induced: i) accumulation in G1-phase (40% increase), ii) reduction in S- and G2-phase (about 50% decrease), iii) a 10-fold increase of cell-death and 2.5-fold increase of apoptosis in sub-G1 phase, iv) a significant increase of NAD+, ATP, and ROS levels, v) a strong inhibition of SIRT2 activity in vitro. NAM significantly delayed tumor growth in vivo (p ≤ 0.0005) and improved survival of melanoma-bearing mice (p ≤ 0.0001). About 3-fold increase (p ≤ 0.05) of Interferon-gamma (IFN-γ) producing cells was observed in NAM treated mice. The plasmatic expression levels of 6 cytokines (namely: Interleukin 5 (IL-5), Eotaxin, Interleukin 12 (p40) (IL12(p40)), Interleukin 3 (IL-3), Interleukin 10 (IL-10) and Regulated on Activation Normal T Expressed and Secreted (RANTES) were significantly changed in the blood of NAM treated mice, suggesting a key role of the immune response. The observed inhibitory effect of NAM on SIRT2 enzymatic activity confirmed previous evidence; we show here that SIRT2 expression is significantly increased in melanoma and inversely related to melanoma-patients survival. Finally, we show for the first time that the expression levels of Niacin receptors HCAR2 and HCAR3 is almost abolished in human melanoma samples. Conclusion NAM shows a relevant anti-melanoma activity in vitro and in vivo and is a suitable candidate for further clinical investigations.
Collapse
|
21
|
Proshkina E, Shaposhnikov M, Moskalev A. Genome-Protecting Compounds as Potential Geroprotectors. Int J Mol Sci 2020; 21:E4484. [PMID: 32599754 PMCID: PMC7350017 DOI: 10.3390/ijms21124484] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/18/2020] [Accepted: 06/19/2020] [Indexed: 02/06/2023] Open
Abstract
Throughout life, organisms are exposed to various exogenous and endogenous factors that cause DNA damages and somatic mutations provoking genomic instability. At a young age, compensatory mechanisms of genome protection are activated to prevent phenotypic and functional changes. However, the increasing stress and age-related deterioration in the functioning of these mechanisms result in damage accumulation, overcoming the functional threshold. This leads to aging and the development of age-related diseases. There are several ways to counteract these changes: 1) prevention of DNA damage through stimulation of antioxidant and detoxification systems, as well as transition metal chelation; 2) regulation of DNA methylation, chromatin structure, non-coding RNA activity and prevention of nuclear architecture alterations; 3) improving DNA damage response and repair; 4) selective removal of damaged non-functional and senescent cells. In the article, we have reviewed data about the effects of various trace elements, vitamins, polyphenols, terpenes, and other phytochemicals, as well as a number of synthetic pharmacological substances in these ways. Most of the compounds demonstrate the geroprotective potential and increase the lifespan in model organisms. However, their genome-protecting effects are non-selective and often are conditioned by hormesis. Consequently, the development of selective drugs targeting genome protection is an advanced direction.
Collapse
Affiliation(s)
- Ekaterina Proshkina
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Centre, Ural Branch, Russian Academy of Sciences, 28 Kommunisticheskaya st., 167982 Syktyvkar, Russia; (E.P.); (M.S.)
| | - Mikhail Shaposhnikov
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Centre, Ural Branch, Russian Academy of Sciences, 28 Kommunisticheskaya st., 167982 Syktyvkar, Russia; (E.P.); (M.S.)
| | - Alexey Moskalev
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Centre, Ural Branch, Russian Academy of Sciences, 28 Kommunisticheskaya st., 167982 Syktyvkar, Russia; (E.P.); (M.S.)
- Pitirim Sorokin Syktyvkar State University, 55 Oktyabrsky prosp., 167001 Syktyvkar, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|