NAD metabolism fuels human and mouse intestinal inflammation

Romana R Gerner, Victoria Klepsch, Sophie Macheiner, Kathrin Arnhard, Timon E Adolph, Christoph Grander, Verena Wieser, Alexandra Pfister, Patrizia Moser, Natascha Hermann-Kleiter, Gottfried Baier, Herbert Oberacher, Herbert Tilg, Alexander R Moschen, Romana R Gerner, Victoria Klepsch, Sophie Macheiner, Kathrin Arnhard, Timon E Adolph, Christoph Grander, Verena Wieser, Alexandra Pfister, Patrizia Moser, Natascha Hermann-Kleiter, Gottfried Baier, Herbert Oberacher, Herbert Tilg, Alexander R Moschen

Abstract

Objective: Nicotinamide phosphoribosyltransferase (NAMPT, also referred to as pre-B cell colony-enhancing factor or visfatin) is critically required for the maintenance of cellular nicotinamide adenine dinucleotide (NAD) supply catalysing the rate-limiting step of the NAD salvage pathway. NAMPT is strongly upregulated in inflammation including IBD and counteracts an increased cellular NAD turnover mediated by NAD-depleting enzymes. These constitute an important mechanistic link between inflammatory, metabolic and transcriptional pathways and NAD metabolism.

Design: We investigated the impact of NAMPT inhibition by the small-molecule inhibitor FK866 in the dextran sulfate sodium (DSS) model of colitis and the azoxymethane/DSS model of colitis-associated cancer. The impact of NAD depletion on differentiation of mouse and human primary monocytes/macrophages was studied in vitro. Finally, we tested the efficacy of FK866 compared with dexamethasone and infliximab in lamina propria mononuclear cells (LPMNC) isolated from patients with IBD.

Results: FK866 ameliorated DSS-induced colitis and suppressed inflammation-associated tumorigenesis in mice. FK866 potently inhibited NAMPT activity as demonstrated by reduced mucosal NAD, resulting in reduced abundances and activities of NAD-dependent enzymes including PARP1, Sirt6 and CD38, reduced nuclear factor kappa B activation, and decreased cellular infiltration by inflammatory monocytes, macrophages and activated T cells. Remarkably, FK866 effectively supressed cytokine release from LPMNCs of patients with IBD. As FK866 was also effective in Rag1-⁄- mice, we mechanistically linked FK866 treatment with altered monocyte/macrophage biology and skewed macrophage polarisation by reducing CD86, CD38, MHC-II and interleukin (IL)-6 and promoting CD206, Egr2 and IL-10.

Conclusion: Our data emphasise the importance of NAD immunometabolism for mucosal immunity and highlight FK866-mediated NAMPT blockade as a promising therapeutic approach in acute intestinal inflammation.

Keywords: Colonic Mucosal Metabolism; Energy Metabolism; Gut Inflammation; IBD Basic Research; Inflammatory Bowel Disease.

Conflict of interest statement

Competing interests: None declared.

© Article author(s) (or their employer(s) unless otherwise stated in the text of the article) 2018. All rights reserved. No commercial use is permitted unless otherwise expressly granted.

Figures

Figure 1
Figure 1
FK866 treatment mitigates clinical, morphological and biochemical parameters of DSS colitis. (A) Colonic Nampt mRNA induction as determined by RT-qPCR (black labelling) is shown together with the weight course (grey symbols and labelling) and NAMPT protein expression using immunoblot analyses along with densitometric quantification during DSS colitis. (B) Comparative NAMPT immunohistochemistry of colons with or without DSS on day 9 of experiments is shown (left panel). NAMPT positivity (red) is observed in IEC and inflammatory cells (15× magnification; scale bars 100 µm). Colonic NAMPT expression of Epcam+epithelial cells and CD45+LPMNC including respective immune cell subsets was analysed by flow cytometry in the steady state and on day 7 of colitis (pie charts). (C) Weight course of vehicle control and FK866-treated mice during DSS colitis. The experimental outline is shown above. (D) Mean colon lengths and faecal Lcn2 of respective groups are shown. (E) Representative H&E stainings of colons along with histological severity scores (5× magnification; scale bars 500 µm). (F) mRNA expression of cytokines and chemokines of mucosal scrapings was determined by RT-qPCR and normalised to ß-actin (day 7 of experiments). n=6–10 per group, five independent experiments. Data represent mean±SEM. *p<0.05; **p<0.01; ***p<0.001; ****p<0.0001; Student’s t-test or one-way analysis of variance followed by Bonferroni’s post hoc corrections when more than three groups analysed. CCL, CC-chemokine ligand; CXCL, C-X-C motif ligand; DSS, dextran sulfate sodium; IEC, intestinal epithelial cells; IL, interleukin; IP, induced protein; Lcn2, lipocalin 2; LPMNC, lamina propria mononuclear cells; Mip, macrophage inflammatory protein; NAMPT, nicotinamide phosphoribosyltransferase; RLU, relative light units; TNFα, tumour necrosis factor alpha.
Figure 2
Figure 2
FK866 treatment is effective independent of T and B cells. (A) Weight course of vehicle control and FK866-treated RAG−/− mice during DSS colitis along with the experimental outline shown above. (B) Representative H&E-stained colonic sections along with histology score of indicated groups (5× magnification; scale bars 500 µm). (C,D) Mean colon lengths and (D) faecal Lcn2 of experimental groups are shown. (E) Colonic scrapings were analysed for mRNA expression of respective cytokines/chemokines on experimental day 7 with or without DSS. n=10 per group, two independent experiments. Data represent mean±SEM. *p<0.05; **p<0.01; ***p<0.001; ****p<0.0001; Student’s t-test or one-way analysis of variance followed by Bonferroni’s post hoc corrections when more than three groups analysed. CCL, CC-chemokine ligand; CXCL, C-X-C motif ligand; DSS, dextran sulfate sodium; IL, interleukin; IP, induced protein; Lcn2, lipocalin 2; Mip, macrophage inflammatory protein; TNFα, tumour necrosis factor alpha.
Figure 3
Figure 3
NAMPT blockage results in a diminished recruitment of inflammatory/activated cells upon intestinal inflammation. (A) Total numbers of colonic infiltrates were quantified by automated cell counting (left). The absolute numbers of CD11b+F4/80hi tissue macrophages, CD11c+MHC-II+-activated dendritic cells, CD11b+Ly6Cloresident monocytes and CD11b+Ly6Chi inflammatory monocytes per colon with or without FK866 during tap water or DSS colitis were analysed by flow cytometry on day 7. (B) On day 7 of colitis, the absolute numbers of CD19+ B cells, CD3+, CD4+ and CD4+CD38+, and CD8+ and CD8+CD38+ T cells per colon of respective groups were analysed by flow cytometry. (C) The mRNA expression of common adhesion molecules was determined by RT-qPCR in mucosal scrapings (n=6–8, two independent experiments). (D) The frequency of Sca1+ stem cell-like cells, CD11b+Ly6Chi inflammatory and CD11b+Ly6Clo resident monocytes of CD45+-gated BM cells was analysed by flow cytometry (n=4–6, two independent experiments). n=4–8 per group, six independent experiments if not otherwise stated. Data represent mean±SEM. *p<0.05; **p<0.01; ***p<0.001; ****p<0.0001; one-way analysis of variance followed by Bonferroni’s post hoc corrections when more than three groups analysed. BM, bone marrow; DSS, dextran sulfate sodium; LPMNC, lamina propria mononuclear cells; NAMPT, nicotinamide phosphoribosyltransferase.
Figure 5
Figure 5
FK866-treated mice are protected in the AOM/DSS model of colitis-associated cancer. (A) The experimental outline along with the weight course is shown. (B,C) Clinical parameters including (B) survival and (C) mean colon lengths of FK866 compared with vehicle-treated mice during the AOM/DSS model. (D) Cell death was evaluated by the TUNEL method. Three crypts/mouse were analysed for TUNEL+cells and expressed as ratio/total IEC count on the crypt axis. (E) The mean tumour numbers and tumour areas of FK866 or vehicle-treated animals were assessed in H&E-stained Swiss roll sections. Dashed, black lines indicate tumour area (5× magnification; scale bars 500 µm). n=12 per group, two independent experiments. Data represent mean±SEM. *p

Figure 6

Therapeutic FK866 treatment is effective…

Figure 6

Therapeutic FK866 treatment is effective on established colitis and modulates proinflammatory cytokine secretion…

Figure 6
Therapeutic FK866 treatment is effective on established colitis and modulates proinflammatory cytokine secretion in human LPMNC from patients with IBD. (A) Histological severity score of mice treated with DSS at 0, 1 and 3 days. (B) FK866 or vehicle treatment was started 3 days after onset of DSS experiments and animals were assessed for clinical parameters including (B) weight loss, (C) colon length and faecal Lcn2 on experimental day 9. (D) Representative H&E-stained colons and respective colitis scores of indicated groups are depicted. (5× magnification; scale bars 500 µm). (E) A schematic flow chart of LPMNC experiments is outlined. (F) Biopsy-derived LPMNCs from patients with CD (upper panel) or UC (lower panel) were incubated with or without infliximab (10 µg/mL), dexamethasone (2 nM) or FK866 (200 nM) and assayed for cytokine release after 18 hours. Vehicle-stimulated cells were set as 100% and data are expressed as % decrease or increase (n=8–10 per group). n=8 per group if not otherwise stated, two independent experiments. Data represent mean±SEM. *p

Figure 4

FK866-induced NAD depletion results in…

Figure 4

FK866-induced NAD depletion results in reduced PARP1 and Sirt6 activity and skews macrophages…

Figure 4
FK866-induced NAD depletion results in reduced PARP1 and Sirt6 activity and skews macrophages towards a less inflammatory phenotype. (A) An experimental outline of the LC-MS/MS approach is shown. Mucosal metabolite concentrations of the NAD salvage pathway along with ATP concentrations of indicated groups during tap water or DSS exposure were determined and normalised to total protein contents. (B,C) Representative PARP1 and Sirt6 immunoblots from mucosal scrapings of indicated groups along with densitometry, and (C) poly(ADP-ribosylation) (PAR) assay or sirtuin deacetylase activity (DAA). (D) Comparative PARP1 and Sirt6 immunohistochemistry of colons with or without FK866 during tap water or DSS exposure on day 7 of experiments is shown (20× magnification; scale bars 50 µm). (E) BMDMs were differentiated for 8 days followed by polarisation with M1 (LPS/IFNγ) or M2 (IL-4) stimuli for 24 hours. Cells were incubated with or without (=ctr) 5 nM FK866 from day 0 (=FK866_d0) or from day 8 (=FK866_d8). On day 9, cell viability was assessed by a LUNA automated cell counter and cells analysed for typical M1 (MHC-II, CD86) and M2 (CD206) surface markers by flow cytometry. n=2–4 per group. mRNA expression of typical M1 (Socs3, Nos2) and M2 (IL-10, Arg1) markers was analysed by quantitative RT-PCR and normalised to HPRT and SNs were assayed for IL-6 and IL-10 by ELISA on day 9. (F) The M1/M2 ratio in the colon of mice with or without FK866 in the steady state and during DSS colitis on day 7 is shown. The frequencies of colonic F4/80+ CD38+-M1 macrophages of FK866 or vehicle-treated mice are depicted in the overlays. n=2–8 per group if not otherwise stated, three independent experiments. Data represent mean±SEM. *p<0.05; **p<0.01; ***p<0.001; Student’s t-test or one-way analysis of variance followed by Bonferroni’s post hoc corrections when more than three groups analysed. BMDM, bone marrow-derived macrophages; DSS, dextran sulfate sodium; HPRT, hypoxanthine-guanine phosphoribosyltransferase; IFNγ, interferon gamma; IL, interleukin; LC-MS/MS, liquid chromatography tandem-mass spectrometry; LPS, lipopolysaccharide; MHC, major histocompatibility complex; NAD, nicotinamide adenine; dinucleotide; NMN, nicotinamide mononucleotide; PAR, poly(ADP-ribose); PARP, poly(ADP-ribose) polymerase; RLU, relative light units; SN, supernatant.
Similar articles
Cited by
References
    1. Kaser A, Zeissig S, Blumberg RS, et al. . Inflammatory bowel disease. Annu Rev Immunol 2010;28:573–621. 10.1146/annurev-immunol-030409-101225 - DOI - PMC - PubMed
    1. Eaden JA, Abrams KR, Mayberry JF. The risk of colorectal cancer in ulcerative colitis: a meta-analysis. Gut 2001;48:526–35. 10.1136/gut.48.4.526 - DOI - PMC - PubMed
    1. Dignass A, Lindsay JO, Sturm A, et al. . Second European evidence-based consensus on the diagnosis and management of ulcerative colitis part 2: current management. J Crohns Colitis 2012;6:991–1030. 10.1016/j.crohns.2012.09.002 - DOI - PubMed
    1. Dignass A, Van Assche G, Lindsay JO, et al. . The second European evidence-based Consensus on the diagnosis and management of Crohn’s disease: Current management. J Crohns Colitis 2010;4:28–62. 10.1016/j.crohns.2009.12.002 - DOI - PubMed
    1. Feagan BG, Sandborn WJ, Gasink C, et al. . Ustekinumab as Induction and Maintenance Therapy for Crohn’s Disease. N Engl J Med 2016;375:1946–60. 10.1056/NEJMoa1602773 - DOI - PubMed
Show all 53 references
Publication types
MeSH terms
Full text links [x]
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 6
Figure 6
Therapeutic FK866 treatment is effective on established colitis and modulates proinflammatory cytokine secretion in human LPMNC from patients with IBD. (A) Histological severity score of mice treated with DSS at 0, 1 and 3 days. (B) FK866 or vehicle treatment was started 3 days after onset of DSS experiments and animals were assessed for clinical parameters including (B) weight loss, (C) colon length and faecal Lcn2 on experimental day 9. (D) Representative H&E-stained colons and respective colitis scores of indicated groups are depicted. (5× magnification; scale bars 500 µm). (E) A schematic flow chart of LPMNC experiments is outlined. (F) Biopsy-derived LPMNCs from patients with CD (upper panel) or UC (lower panel) were incubated with or without infliximab (10 µg/mL), dexamethasone (2 nM) or FK866 (200 nM) and assayed for cytokine release after 18 hours. Vehicle-stimulated cells were set as 100% and data are expressed as % decrease or increase (n=8–10 per group). n=8 per group if not otherwise stated, two independent experiments. Data represent mean±SEM. *p

Figure 4

FK866-induced NAD depletion results in…

Figure 4

FK866-induced NAD depletion results in reduced PARP1 and Sirt6 activity and skews macrophages…

Figure 4
FK866-induced NAD depletion results in reduced PARP1 and Sirt6 activity and skews macrophages towards a less inflammatory phenotype. (A) An experimental outline of the LC-MS/MS approach is shown. Mucosal metabolite concentrations of the NAD salvage pathway along with ATP concentrations of indicated groups during tap water or DSS exposure were determined and normalised to total protein contents. (B,C) Representative PARP1 and Sirt6 immunoblots from mucosal scrapings of indicated groups along with densitometry, and (C) poly(ADP-ribosylation) (PAR) assay or sirtuin deacetylase activity (DAA). (D) Comparative PARP1 and Sirt6 immunohistochemistry of colons with or without FK866 during tap water or DSS exposure on day 7 of experiments is shown (20× magnification; scale bars 50 µm). (E) BMDMs were differentiated for 8 days followed by polarisation with M1 (LPS/IFNγ) or M2 (IL-4) stimuli for 24 hours. Cells were incubated with or without (=ctr) 5 nM FK866 from day 0 (=FK866_d0) or from day 8 (=FK866_d8). On day 9, cell viability was assessed by a LUNA automated cell counter and cells analysed for typical M1 (MHC-II, CD86) and M2 (CD206) surface markers by flow cytometry. n=2–4 per group. mRNA expression of typical M1 (Socs3, Nos2) and M2 (IL-10, Arg1) markers was analysed by quantitative RT-PCR and normalised to HPRT and SNs were assayed for IL-6 and IL-10 by ELISA on day 9. (F) The M1/M2 ratio in the colon of mice with or without FK866 in the steady state and during DSS colitis on day 7 is shown. The frequencies of colonic F4/80+ CD38+-M1 macrophages of FK866 or vehicle-treated mice are depicted in the overlays. n=2–8 per group if not otherwise stated, three independent experiments. Data represent mean±SEM. *p<0.05; **p<0.01; ***p<0.001; Student’s t-test or one-way analysis of variance followed by Bonferroni’s post hoc corrections when more than three groups analysed. BMDM, bone marrow-derived macrophages; DSS, dextran sulfate sodium; HPRT, hypoxanthine-guanine phosphoribosyltransferase; IFNγ, interferon gamma; IL, interleukin; LC-MS/MS, liquid chromatography tandem-mass spectrometry; LPS, lipopolysaccharide; MHC, major histocompatibility complex; NAD, nicotinamide adenine; dinucleotide; NMN, nicotinamide mononucleotide; PAR, poly(ADP-ribose); PARP, poly(ADP-ribose) polymerase; RLU, relative light units; SN, supernatant.
Figure 4
Figure 4
FK866-induced NAD depletion results in reduced PARP1 and Sirt6 activity and skews macrophages towards a less inflammatory phenotype. (A) An experimental outline of the LC-MS/MS approach is shown. Mucosal metabolite concentrations of the NAD salvage pathway along with ATP concentrations of indicated groups during tap water or DSS exposure were determined and normalised to total protein contents. (B,C) Representative PARP1 and Sirt6 immunoblots from mucosal scrapings of indicated groups along with densitometry, and (C) poly(ADP-ribosylation) (PAR) assay or sirtuin deacetylase activity (DAA). (D) Comparative PARP1 and Sirt6 immunohistochemistry of colons with or without FK866 during tap water or DSS exposure on day 7 of experiments is shown (20× magnification; scale bars 50 µm). (E) BMDMs were differentiated for 8 days followed by polarisation with M1 (LPS/IFNγ) or M2 (IL-4) stimuli for 24 hours. Cells were incubated with or without (=ctr) 5 nM FK866 from day 0 (=FK866_d0) or from day 8 (=FK866_d8). On day 9, cell viability was assessed by a LUNA automated cell counter and cells analysed for typical M1 (MHC-II, CD86) and M2 (CD206) surface markers by flow cytometry. n=2–4 per group. mRNA expression of typical M1 (Socs3, Nos2) and M2 (IL-10, Arg1) markers was analysed by quantitative RT-PCR and normalised to HPRT and SNs were assayed for IL-6 and IL-10 by ELISA on day 9. (F) The M1/M2 ratio in the colon of mice with or without FK866 in the steady state and during DSS colitis on day 7 is shown. The frequencies of colonic F4/80+ CD38+-M1 macrophages of FK866 or vehicle-treated mice are depicted in the overlays. n=2–8 per group if not otherwise stated, three independent experiments. Data represent mean±SEM. *p<0.05; **p<0.01; ***p<0.001; Student’s t-test or one-way analysis of variance followed by Bonferroni’s post hoc corrections when more than three groups analysed. BMDM, bone marrow-derived macrophages; DSS, dextran sulfate sodium; HPRT, hypoxanthine-guanine phosphoribosyltransferase; IFNγ, interferon gamma; IL, interleukin; LC-MS/MS, liquid chromatography tandem-mass spectrometry; LPS, lipopolysaccharide; MHC, major histocompatibility complex; NAD, nicotinamide adenine; dinucleotide; NMN, nicotinamide mononucleotide; PAR, poly(ADP-ribose); PARP, poly(ADP-ribose) polymerase; RLU, relative light units; SN, supernatant.

References

    1. Kaser A, Zeissig S, Blumberg RS, et al. . Inflammatory bowel disease. Annu Rev Immunol 2010;28:573–621. 10.1146/annurev-immunol-030409-101225
    1. Eaden JA, Abrams KR, Mayberry JF. The risk of colorectal cancer in ulcerative colitis: a meta-analysis. Gut 2001;48:526–35. 10.1136/gut.48.4.526
    1. Dignass A, Lindsay JO, Sturm A, et al. . Second European evidence-based consensus on the diagnosis and management of ulcerative colitis part 2: current management. J Crohns Colitis 2012;6:991–1030. 10.1016/j.crohns.2012.09.002
    1. Dignass A, Van Assche G, Lindsay JO, et al. . The second European evidence-based Consensus on the diagnosis and management of Crohn’s disease: Current management. J Crohns Colitis 2010;4:28–62. 10.1016/j.crohns.2009.12.002
    1. Feagan BG, Sandborn WJ, Gasink C, et al. . Ustekinumab as Induction and Maintenance Therapy for Crohn’s Disease. N Engl J Med 2016;375:1946–60. 10.1056/NEJMoa1602773
    1. Feagan BG, Rutgeerts P, Sands BE, et al. . Vedolizumab as induction and maintenance therapy for ulcerative colitis. N Engl J Med 2013;369:699–710. 10.1056/NEJMoa1215734
    1. Allez M, Karmiris K, Louis E, et al. . Report of the ECCO pathogenesis workshop on anti-TNF therapy failures in inflammatory bowel diseases: definitions, frequency and pharmacological aspects. J Crohns Colitis 2010;4:355–66. 10.1016/j.crohns.2010.04.004
    1. Sands BE, Feagan BG, Rutgeerts P, et al. . Effects of vedolizumab induction therapy for patients with Crohn’s disease in whom tumor necrosis factor antagonist treatment failed. Gastroenterology 2014;147:618–27. 10.1053/j.gastro.2014.05.008
    1. Garedew A, Henderson SO, Moncada S. Activated macrophages utilize glycolytic ATP to maintain mitochondrial membrane potential and prevent apoptotic cell death. Cell Death Differ 2010;17:1540–50. 10.1038/cdd.2010.27
    1. Krawczyk CM, Holowka T, Sun J, et al. . Toll-like receptor-induced changes in glycolytic metabolism regulate dendritic cell activation. Blood 2010;115:4742–9. 10.1182/blood-2009-10-249540
    1. Kominsky DJ, Campbell EL, Colgan SP. Metabolic shifts in immunity and inflammation. J Immunol 2010;184:4062–8. 10.4049/jimmunol.0903002
    1. Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 2009;324:1029–33. 10.1126/science.1160809
    1. Schreiber V, Dantzer F, Ame JC, et al. . Poly(ADP-ribose): novel functions for an old molecule. Nat Rev Mol Cell Biol 2006;7:517–28. 10.1038/nrm1963
    1. Michan S, Sinclair D. Sirtuins in mammals: insights into their biological function. Biochem J 2007;404:1–13. 10.1042/BJ20070140
    1. Partida-Sanchez S, Gasser A, Fliegert R, et al. . Chemotaxis of mouse bone marrow neutrophils and dendritic cells is controlled by adp-ribose, the major product generated by the CD38 enzyme reaction. J Immunol 2007;179:7827–39. 10.4049/jimmunol.179.11.7827
    1. Cantó C, Menzies KJ, Auwerx J. NAD(+) Metabolism and the Control of Energy Homeostasis: A Balancing Act between Mitochondria and the Nucleus. Cell Metab 2015;22:31–53. 10.1016/j.cmet.2015.05.023
    1. Rongvaux A, Shea RJ, Mulks MH, et al. . Pre-B-cell colony-enhancing factor, whose expression is up-regulated in activated lymphocytes, is a nicotinamide phosphoribosyltransferase, a cytosolic enzyme involved in NAD biosynthesis. Eur J Immunol 2002;32:3225–34. 10.1002/1521-4141(200211)32:11<3225::AID-IMMU3225>;2-L
    1. Jia SH, Li Y, Parodo J, et al. . Pre-B cell colony-enhancing factor inhibits neutrophil apoptosis in experimental inflammation and clinical sepsis. J Clin Invest 2004;113:1318–27. 10.1172/JCI19930
    1. Meier FM, Frommer KW, Peters MA, et al. . Visfatin/pre-B-cell colony-enhancing factor (PBEF), a proinflammatory and cell motility-changing factor in rheumatoid arthritis. J Biol Chem 2012;287:28378–85. 10.1074/jbc.M111.312884
    1. El-Mesallamy HO, Kassem DH, El-Demerdash E, et al. . Vaspin and visfatin/Nampt are interesting interrelated adipokines playing a role in the pathogenesis of type 2 diabetes mellitus. Metabolism 2011;60:63–70. 10.1016/j.metabol.2010.04.008
    1. Galli U, Travelli C, Massarotti A, et al. . Medicinal chemistry of nicotinamide phosphoribosyltransferase (NAMPT) inhibitors. J Med Chem 2013;56:6279–96. 10.1021/jm4001049
    1. Moschen AR, Kaser A, Enrich B, et al. . Visfatin, an adipocytokine with proinflammatory and immunomodulating properties. J Immunol 2007;178:1748–58. 10.4049/jimmunol.178.3.1748
    1. Starr AE, Deeke SA, Ning Z, et al. . Proteomic analysis of ascending colon biopsies from a paediatric inflammatory bowel disease inception cohort identifies protein biomarkers that differentiate Crohn’s disease from UC. Gut 2017;66 10.1136/gutjnl-2015-310705
    1. Hasmann M, Schemainda I. FK866, a highly specific noncompetitive inhibitor of nicotinamide phosphoribosyltransferase, represents a novel mechanism for induction of tumor cell apoptosis. Cancer Res 2003;63:7436–42.
    1. Moschen AR, Gerner R, Schroll A, et al. . A key role for Pre-B cell colony-enhancing factor in experimental hepatitis. Hepatology 2011;54:675–86. 10.1002/hep.24416
    1. Bruzzone S, Fruscione F, Morando S, et al. . Catastrophic NAD+ depletion in activated T lymphocytes through Nampt inhibition reduces demyelination and disability in EAE. PLoS One 2009;4:e7897 10.1371/journal.pone.0007897
    1. Oikonomou KA, Kapsoritakis AN, Theodoridou C, et al. . Neutrophil gelatinase-associated lipocalin (NGAL) in inflammatory bowel disease: association with pathophysiology of inflammation, established markers, and disease activity. J Gastroenterol 2012;47:519–30. 10.1007/s00535-011-0516-5
    1. Moschen AR, Gerner RR, Wang J, et al. . Lipocalin 2 Protects from Inflammation and Tumorigenesis Associated with Gut Microbiota Alterations. Cell Host Microbe 2016;19:455–69. 10.1016/j.chom.2016.03.007
    1. Rongvaux A, Galli M, Denanglaire S, et al. . Nicotinamide phosphoribosyl transferase/pre-B cell colony-enhancing factor/visfatin is required for lymphocyte development and cellular resistance to genotoxic stress. J Immunol 2008;181:4685–95. 10.4049/jimmunol.181.7.4685
    1. Shmuel-Galia L, Aychek T, Fink A, et al. . Neutralization of pro-inflammatory monocytes by targeting TLR2 dimerization ameliorates colitis. Embo J 2016;35:685–98.
    1. Bain CC, Scott CL, Uronen-Hansson H, et al. . Resident and pro-inflammatory macrophages in the colon represent alternative context-dependent fates of the same Ly6Chi monocyte precursors. Mucosal Immunol 2013;6:498–510. 10.1038/mi.2012.89
    1. Busso N, Karababa M, Nobile M, et al. . Pharmacological inhibition of nicotinamide phosphoribosyltransferase/visfatin enzymatic activity identifies a new inflammatory pathway linked to NAD. PLoS One 2008;3:e2267 10.1371/journal.pone.0002267
    1. Hassa PO, Haenni SS, Buerki C, et al. . Acetylation of poly(ADP-ribose) polymerase-1 by p300/CREB-binding protein regulates coactivation of NF-kappaB-dependent transcription. J Biol Chem 2005;280:40450–64. 10.1074/jbc.M507553200
    1. Kawahara TL, Michishita E, Adler AS, et al. . SIRT6 links histone H3 lysine 9 deacetylation to NF-kappaB-dependent gene expression and organismal life span. Cell 2009;136:62–74. 10.1016/j.cell.2008.10.052
    1. Sica A, Schioppa T, Mantovani A, et al. . Tumour-associated macrophages are a distinct M2 polarised population promoting tumour progression: potential targets of anti-cancer therapy. Eur J Cancer 2006;42:717–27. 10.1016/j.ejca.2006.01.003
    1. Moschen AR, Gerner RR, Tilg H. Pre-B cell colony enhancing factor/NAMPT/visfatin in inflammation and obesity-related disorders. Curr Pharm Des 2010;16:1913–20. 10.2174/138161210791208947
    1. Samal B, Sun Y, Stearns G, et al. . Cloning and characterization of the cDNA encoding a novel human pre-B-cell colony-enhancing factor. Mol Cell Biol 1994;14:1431–7. 10.1128/MCB.14.2.1431
    1. Li Y, Zhang Y, Dorweiler B, et al. . Extracellular Nampt promotes macrophage survival via a nonenzymatic interleukin-6/STAT3 signaling mechanism. J Biol Chem 2008;283:34833–43. 10.1074/jbc.M805866200
    1. Van Gool F, Gallí M, Gueydan C, et al. . Intracellular NAD levels regulate tumor necrosis factor protein synthesis in a sirtuin-dependent manner. Nat Med 2009;15:206–10. 10.1038/nm.1906
    1. Chiarugi A, Dölle C, Felici R, et al. . The NAD metabolome--a key determinant of cancer cell biology. Nat Rev Cancer 2012;12:741–52. 10.1038/nrc3340
    1. Carter L, Morgan R, Lesch C, et al. . Targeting immune cell metabolism: LYC-30937, a novel therapeutic approach for inflammatory bowel disease. Journal of Crohns & Colitis 2016;10:S38–S9.
    1. Kitani T, Okuno S, Fujisawa H. Growth phase-dependent changes in the subcellular localization of pre-B-cell colony-enhancing factor. FEBS Lett 2003;544:74–8. 10.1016/S0014-5793(03)00476-9
    1. Larmonier CB, Shehab KW, Laubitz D, et al. . Transcriptional Reprogramming and Resistance to Colonic Mucosal Injury in Poly(ADP-ribose) Polymerase 1 (PARP1)-deficient Mice. J Biol Chem 2016;291:8918–30. 10.1074/jbc.M116.714386
    1. Jagtap P, Szabó C, Poly SC. Poly(ADP-ribose) polymerase and the therapeutic effects of its inhibitors. Nat Rev Drug Discov 2005;4:421–40. 10.1038/nrd1718
    1. Boughton-Smith NK, Evans SM, Hawkey CJ, et al. . Nitric oxide synthase activity in ulcerative colitis and Crohn’s disease. Lancet 1993;342:338–e2. 10.1016/0140-6736(93)91476-3
    1. Singer II, Kawka DW, Scott S, et al. . Expression of inducible nitric oxide synthase and nitrotyrosine in colonic epithelium in inflammatory bowel disease. Gastroenterology 1996;111:871–85. 10.1016/S0016-5085(96)70055-0
    1. Brunyanszki A, Olah G, Coletta C, et al. . Regulation of mitochondrial poly(ADP-Ribose) polymerase activation by the β-adrenoceptor/cAMP/protein kinase A axis during oxidative stress. Mol Pharmacol 2014;86:450–62. 10.1124/mol.114.094318
    1. Chen X, Lu Y, Zhang Z, et al. . Intercellular interplay between Sirt1 signalling and cell metabolism in immune cell biology. Immunology 2015;145:455–67. 10.1111/imm.12473
    1. Sandoval-Montes C, Santos-Argumedo L. CD38 is expressed selectively during the activation of a subset of mature T cells with reduced proliferation but improved potential to produce cytokines. J Leukoc Biol 2005;77:513–21. 10.1189/jlb.0404262
    1. Abraham C, Cho JH. Inflammatory bowel disease. N Engl J Med 2009;361:2066–78. 10.1056/NEJMra0804647
    1. Biswas SK, Mantovani A. Orchestration of metabolism by macrophages. Cell Metab 2012;15:432–7. 10.1016/j.cmet.2011.11.013
    1. Venter G, Oerlemans FT, Willemse M, et al. . NAMPT-mediated salvage synthesis of NAD+ controls morphofunctional changes of macrophages. PLoS One 2014;9:e97378 10.1371/journal.pone.0097378
    1. Holen K, Saltz LB, Hollywood E, et al. . The pharmacokinetics, toxicities, and biologic effects of FK866, a nicotinamide adenine dinucleotide biosynthesis inhibitor. Invest New Drugs 2008;26:45–51. 10.1007/s10637-007-9083-2

Source: PubMed

3
Subscribe