Re-equilibration of imbalanced NAD metabolism ameliorates the impact of telomere dysfunction

Chongkui Sun, Kun Wang, Amanda J Stock, Yi Gong, Tyler G Demarest, Beimeng Yang, Neelam Giri, Lea Harrington, Blanche P Alter, Sharon A Savage, Vilhelm A Bohr, Yie Liu, Chongkui Sun, Kun Wang, Amanda J Stock, Yi Gong, Tyler G Demarest, Beimeng Yang, Neelam Giri, Lea Harrington, Blanche P Alter, Sharon A Savage, Vilhelm A Bohr, Yie Liu

Abstract

Short telomeres are a principal defining feature of telomere biology disorders, such as dyskeratosis congenita (DC), for which there are no effective treatments. Here, we report that primary fibroblasts from DC patients and late generation telomerase knockout mice display lower nicotinamide adenine dinucleotide (NAD) levels, and an imbalance in the NAD metabolome that includes elevated CD38 NADase and reduced poly(ADP-ribose) polymerase and SIRT1 activities, respectively, affecting many associated biological pathways. Supplementation with the NAD precursor, nicotinamide riboside, and CD38 inhibition improved NAD homeostasis, thereby alleviating telomere damage, defective mitochondrial biosynthesis and clearance, cell growth retardation, and cellular senescence of DC fibroblasts. These findings reveal a direct, underlying role of NAD dysregulation when telomeres are short and underscore its relevance to the pathophysiology and interventions of human telomere-driven diseases.

Keywords: CD38 NADase; NAD metabolism; mitochondrial impairment; replicative senescence; telomere biology disorders.

Conflict of interest statement

The authors declare that they have no conflict of interest.

© 2020 The Authors. Published under the terms of the CC BY NC ND 4.0 license.

Figures

Figure 1. DC patient fibroblasts exhibit defects…
Figure 1. DC patient fibroblasts exhibit defects in NAD metabolism and NAD‐dependent PARylation and SIRT1 deacetylation activity
  1. A, B

    Intracellular NAD levels and NAD/NADH ratio in DC and age‐matched healthy control fibroblasts. All values are presented as mean ± SD of four and eight replicates in (A) and (B), respectively. Student's t‐test was performed on DC cells vs controls.

  2. C

    An overview of NAD metabolism. The activity of NAD‐consuming enzymes, such as PARPs, SIRTs, or CD38 consumes NAD and results in NAM production. The NAD biosynthesizing enzymes NMNATs and NAMPT recycle NAM back to NMN and then NMN to NAD, respectively. NR is converted to NMN, followed by conversion of NMN to NAD.

  3. D, E

    Immunoblots of the expression of NAD synthesis and consuming proteins, and their activities in DC and control cells. Protein levels are normalized to GAPDH. Quantification values are presented as mean ± SD of four controls vs five DC samples as shown in (D). Student's t‐test was performed on DC cells vs controls. Con: control fibroblasts. Irrelevant intervening lanes in (D) have been removed for clarity (full blots are available online as Source Data).

  4. F

    Effects of NR and NAM supplementation (24 h) on the NAD levels in DC fibroblasts. All values are presented as mean ± SD of four replicates. One‐way ANOVA was performed on DC cells in indicated conditions.

Source data are available online for this figure.
Figure 2. CD38 depletion ameliorates dysregulated NAD…
Figure 2. CD38 depletion ameliorates dysregulated NAD metabolism in DC cells
  1. A

    Quantitative RT‐PCR and immunoblots of the expression of CD38 and SARM1 in DC and age‐matched health control fibroblasts. All values are presented as mean ± SD of three replicates in quantitative RT‐PCR. Protein levels are normalized to GAPDH, and mean (± SD) quantification values of four controls vs five DC samples are shown. Student's t‐test was performed on DC cells vs controls. Irrelevant intervening lanes have been removed for clarity (full blots are available online as Source Data).

  2. B

    Immunoblots and quantification of indicated proteins in DC fibroblasts treated with DMSO and with 2 μM ATM inhibitor, KU‐55933 for 48 h. Mean (± SD) quantification values of three DC lines with and without ATMi treatment are shown. P values on the basis of Student's t‐test.

  3. C

    The efficacy of CD38 shRNA (sh‐1 and sh‐2) was verified by quantitative RT‐PCR in DC fibroblasts and by RT‐PCR and immunoblots in A549 cells. The relative CD38 mRNA values in the CD38 knockdown cells were normalized to the scrambled shRNA. All values are presented as mean ± SD of three replicates. Student's t‐test was performed on DC and A549 cells in indicated conditions.

  4. D, E

    Intracellular NAD levels and immunoblots of the expression of indicated proteins in the scramble and CD38 knockdown DC fibroblasts. The CD38 knockdown DC fibroblasts were treated with the PARP1 inhibitor Olaparib (400 nM) and the SIRT1 inhibitor EX 527 (1 μM) for 24 h. All values are presented as mean ± SD of four replicates. One‐way ANOVA was performed on DC cells in indicated conditions.

Source data are available online for this figure.
Figure 3. G3 Tert −/− mice display…
Figure 3. G3 Tert −/− mice display defects in the NAD metabolism
  1. A, B

    Intracellular NAD levels and immunoblots of the expression of NAD consuming‐related proteins and their activities in brain tissues from 6‐month‐old G1 and G3 Tert/ female mice. The CD38 antibody was validated using the CD38/ mouse brain tissues.

  2. C

    Quantification of immunoblots from the indicated proteins in (B).

  3. D

    NADase activity in G1 and G3 Tert/ mouse brain lysate was determined at various time points during the reaction. NADase activity was not detectable in the extracts treated with the CD38 inhibitor, 78c, at a concentration of 50 nM.

Data information: All values are presented as mean ± SD of G1 Tert/ mouse brain tissues vs G3 Tert/ mouse brain tissues. n = 4 in each group. P values on the basis of Student's t‐test.
Figure 4. NAD intervention impacts mitochondrial parameters…
Figure 4. NAD intervention impacts mitochondrial parameters of DC fibroblasts
  1. A

    Representative immunoblots of the expression of PGC1‐1α in DC cells treated with vehicle or 3 mM NR. Quantification of the indicated proteins is from three immunoblots.

  2. B–D

    Cellular and mitochondrial ROS in DC and age‐matched healthy control cells supplemented with vehicle and NR (B, C) and in the scramble and CD38 knockdown DC cells (D) were measured by flow cytometry from three replicates.

  3. E

    Representative images captured by transmission electron microscopy of DC1 cells treated with vehicle or NR. White arrows: mitochondria. Yellow arrow: autophagosome‐like structures with engulfed mitochondria. Enlarged image within white frame is shown.

  4. F–H

    Quantifications of percentage of damaged mitochondria per cell (F), mitochondrial length (G), and mitochondrial diameter (H). A minimum of 200 mitochondria counted per group.

Data information: All data are represented as mean ± SD. Student's t‐test was performed for individual pairs in indicated conditions. Con: control.
Figure 5. Impaired mitophagy process in DC…
Figure 5. Impaired mitophagy process in DC fibroblasts is improved by NR supplementation
  1. Representative images of mitophagy showing co‐localization between the mitophagy dye (red) and the lysosome dye (green) in DC and control fibroblasts treated with vehicle or 3 mM NR. White arrows: mitophagy.

  2. The mean value of fluorescence intensity/cells in each image was scored. At least 15 images (˜ 200 cells) were counted per group. The relative values in each group were normalized to Con1.

  3. Representative immunoblots of the expression of PINK1 and PARKIN in DC cells treated with vehicle or 3 mM NR. Quantification of immunoblots from the indicated proteins is from three replicates.

Data information: All values are represented as mean ± SD. One‐way ANOVA (B) and Student's t‐test (C) were performed for each individual pair. Con: age‐matched healthy controls.
Figure 6. NR supplementation reduces telomeric oxidative…
Figure 6. NR supplementation reduces telomeric oxidative DNA lesions and TIF formation in DC cells
  1. A

    PCR amplification efficiency at the telomere in the mock‐ and FPG‐treated DC and age‐matched healthy control fibroblasts supplemented with vehicle or 3 mM NR. All values are presented as mean ± SD of three replicates. The relative telomere PCR amplification in each sample was normalized to the 36B4 reference gene.

  2. B

    Telomere restriction fragment analysis in DC and control fibroblasts treated with vehicle or 3 mM NR. Genomic DNA was isolated from the indicated cell lines at 0 or 14 days, with or without 3 mM NR treatment. At left, DNA molecular mass markers, in kilobase pairs (Kb).

  3. C, D

    Representative images of γH2AX (green), telomere immuno‐FISH (red) in DC cells treated with vehicle or 3 mM NR. White arrows: co‐localization of γH2AX foci and telomeric DNA (or TIF). Enlarged views of co‐localizing foci are shown at the right panel and in Fig EV1. Percentage of DC and control cells with indicated TIFs per nuclei. Approximately 100 cells per condition were scored.

Data information: All values are represented as mean ± SD. P values were calculated using One‐way ANOVA (A, D). Con: age‐matched healthy controls.
Figure EV1. TIF formation in DC cells
Figure EV1. TIF formation in DC cells
Enlarged view of the upper right panel of Fig 6C, showing a representative image with γH2AX (green) and telomere immuno‐FISH (red) in a DC cell. White arrows: co‐localization of γH2AX foci and telomeric DNA (or TIF). Enlarged view of co‐localizing foci is shown at the right panel.
Figure 7. NAD intervention improves the proliferative…
Figure 7. NAD intervention improves the proliferative capacity and suppresses cellular senescence and SASP in DC fibroblasts
  1. A

    Cumulative population doubling analysis of the proliferation of representative scramble and CD38 knockdown DC fibroblasts or DC fibroblasts treated with vehicle or 3 mM NR. Each data point is represented as mean ± SD of three replicates.

  2. B, C

    Representative images of BrdU (red) and DAPI (blue) staining of DC and age‐matched healthy control fibroblasts treated with vehicle or NR. Percentage of BrdU‐positive cells per condition. Each dot represents the percentage of cells with BrdU staining per image. Approximately 400 cells were counted per condition. All values are presented as mean ± SD.

  3. D, E

    Representative images of SPiDER‐β‐gal (green) and DAPI (blue) staining of DC and age‐matched healthy control cells treated with vehicle or 3 mM NR. Percentage of SA‐β‐gal-positive cells per condition. Each dot represents the percentage of cells with SA‐β‐gal staining per image. Approximately 400 cells were counted per condition. All values are presented as mean ± SD.

  4. F

    The levels of IL‐6, IL‐8, and MCP‐1 in both supernatant and cell lysate of indicated DC fibroblasts treated with vehicle or 3 mM NR. All values are represented as mean ± SD of three replicates.

  5. G, H

    Representative immunoblots of p21 and p16 in DC fibroblasts treated with vehicle or 3 mM NR. Quantification of the indicated proteins is presented as mean ± SD from three immunoblots.

Data information: P values were calculated using one‐way ANOVA for multiple comparisons (C) and Student's t‐test between two groups (C, E, F, and H). Con: control.

References

    1. d'Adda di Fagagna F, Reaper PM, Clay‐Farrace L, Fiegler H, Carr P, Von Zglinicki T, Saretzki G, Carter NP, Jackson SP (2003) A DNA damage checkpoint response in telomere‐initiated senescence. Nature 426: 194–198
    1. Aksoy P, White TA, Thompson M, Chini EN (2006) Regulation of intracellular levels of NAD: a novel role for CD38. Biochem Biophys Res Commun 345: 1386–1392
    1. Alter BP, Giri N, Savage SA, Rosenberg PS (2018) Cancer in the National Cancer Institute inherited bone marrow failure syndrome cohort after fifteen years of follow‐up. Haematologica 103: 30–39
    1. Amano H, Chaudhury A, Rodriguez‐Aguayo C, Lu L, Akhanov V, Catic A, Popov YV, Verdin E, Johnson H, Stossi F et al (2019) Telomere dysfunction induces sirtuin repression that drives telomere‐dependent disease. Cell Metab 29: 1274–1290
    1. Armanios M (2013) Telomeres and age‐related disease: how telomere biology informs clinical paradigms. J Clin Invest 123: 996–1002
    1. Bertuch AA (2016) The molecular genetics of the telomere biology disorders. RNA Biol 13: 696–706
    1. Bieganowski P, Brenner C (2004) Discoveries of nicotinamide riboside as a nutrient and conserved NRK genes establish a Preiss‐Handler independent route to NAD+ in fungi and humans. Cell 117: 495–502
    1. Bischoff DS, Makhijani NS, Yamaguchi DT (2012) Constitutive expression of human telomerase enhances the proliferation potential of human mesenchymal stem cells. Biores Open Access 1: 273–279
    1. Brind'Amour J, Lansdorp PM (2011) Analysis of repetitive DNA in chromosomes by flow cytometry. Nat Methods 8: 484–486
    1. Camacho‐Pereira J, Tarrago MG, Chini CCS, Nin V, Escande C, Warner GM, Puranik AS, Schoon RA, Reid JM, Galina A et al (2016) CD38 dictates age‐related NAD decline and mitochondrial dysfunction through an SIRT3‐dependent mechanism. Cell Metab 23: 1127–1139
    1. Canto C, Menzies KJ, Auwerx J (2015) NAD(+) metabolism and the control of energy homeostasis: a balancing act between mitochondria and the nucleus. Cell Metab 22: 31–53
    1. Cawthon RM (2002) Telomere measurement by quantitative PCR. Nucleic Acids Res 30: e47
    1. Chini CCS, Tarrago MG, Chini EN (2017) NAD and the aging process: role in life, death and everything in between. Mol Cell Endocrinol 455: 62–74
    1. Chini C, Hogan KA, Warner GM, Tarrago MG, Peclat TR, Tchkonia T, Kirkland JL, Chini E (2019) The NADase CD38 is induced by factors secreted from senescent cells providing a potential link between senescence and age‐related cellular NAD(+) decline. Biochem Biophys Res Commun 513: 486–493
    1. Coppe JP, Desprez PY, Krtolica A, Campisi J (2010) The senescence‐associated secretory phenotype: the dark side of tumor suppression. Annu Rev Pathol 5: 99–118
    1. Covarrubias AJ, Kale A, Perrone R, Lopez‐Dominguez JA, Pisco AO, Kasler HG, Schmidt MS, Wiley CD, Iyer SS, Basisty N et al (2019) Aging‐related inflammation driven by cellular senescence enhances NAD consumption via activation of CD38+ pro‐inflammatory macrophages. bioRXiv 10.1101/609438 [PREPRINT]
    1. Dokal I, Vulliamy T, Mason P, Bessler M (2015) Clinical utility gene card for: dyskeratosis congenita ‐ update 2015. Eur J Hum Genet 23: 558
    1. Eisemann T, Pascal JM (2020) Poly(ADP‐ribose) polymerase enzymes and the maintenance of genome integrity. Cell Mol Life Sci 77: 19–33
    1. Erdmann N, Liu Y, Harrington L (2004) Distinct dosage requirements for the maintenance of long and short telomeres in mTert heterozygous mice. Proc Natl Acad Sci USA 101: 6080–6085
    1. Fang EF, Scheibye‐Knudsen M, Brace LE, Kassahun H, SenGupta T, Nilsen H, Mitchell JR, Croteau DL, Bohr VA (2014) Defective mitophagy in XPA via PARP‐1 hyperactivation and NAD(+)/SIRT1 reduction. Cell 157: 882–896
    1. Fang EF, Kassahun H, Croteau DL, Scheibye‐Knudsen M, Marosi K, Lu H, Shamanna RA, Kalyanasundaram S, Bollineni RC, Wilson MA et al (2016) NAD+ replenishment improves lifespan and healthspan in Ataxia telangiectasia models via mitophagy and DNA repair. Cell Metab 24: 566–581
    1. Fang EF, Lautrup S, Hou Y, Demarest TG, Croteau DL, Mattson MP, Bohr VA (2017) NAD(+) in aging: molecular mechanisms and translational implications. Trends Mol Med 23: 899–916
    1. Fernandez‐Marcos PJ, Auwerx J (2011) Regulation of PGC‐1alpha, a nodal regulator of mitochondrial biogenesis. Am J Clin Nutr 93: 884S–890
    1. Frederick DW, Loro E, Liu L, Davila A Jr, Chellappa K, Silverman IM, Quinn WJ 3rd, Gosai SJ, Tichy ED, Davis JG et al (2016) Loss of NAD homeostasis leads to progressive and reversible degeneration of skeletal muscle. Cell Metab 24: 269–282
    1. Gomes AP, Price NL, Ling AJ, Moslehi JJ, Montgomery MK, Rajman L, White JP, Teodoro JS, Wrann CD, Hubbard BP et al (2013) Declining NAD(+) induces a pseudohypoxic state disrupting nuclear‐mitochondrial communication during aging. Cell 155: 1624–1638
    1. Jang SY, Kang HT, Hwang ES (2012) Nicotinamide‐induced mitophagy: event mediated by high NAD+/NADH ratio and SIRT1 protein activation. J Biol Chem 287: 19304–19314
    1. Lin SC, Hardie DG (2018) AMPK: sensing glucose as well as cellular energy status. Cell Metab 27: 299–313
    1. Liu Y, Snow BE, Hande MP, Yeung D, Erdmann NJ, Wakeham A, Itie A, Siderovski DP, Lansdorp PM, Robinson MO et al (2000) The telomerase reverse transcriptase is limiting and necessary for telomerase function in vivo . Curr Biol 10: 1459–1462
    1. Livak KJ, Schmittgen TD (2001) Analysis of relative gene expression data using real‐time quantitative PCR and the 2(‐Delta Delta C(T)) Method. Methods 25: 402–408
    1. Lopez‐Otin C, Blasco MA, Partridge L, Serrano M, Kroemer G (2013) The hallmarks of aging. Cell 153: 1194–1217
    1. Ma C, Pi C, Yang Y, Lin L, Shi Y, Li Y, Li Y, He X (2017) Nampt expression decreases age‐related senescence in rat bone marrow mesenchymal stem cells by targeting Sirt1. PLoS ONE 12: e0170930
    1. Mendelsohn AR, Larrick JW (2017) The NAD+/PARP1/SIRT1 axis in aging. Rejuvenation Res 20: 244–247
    1. Mendelsohn AR, Larrick JW (2019) Interacting NAD(+) and cell senescence pathways complicate antiaging therapies. Rejuvenation Res 22: 261–266
    1. Mills KF, Yoshida S, Stein LR, Grozio A, Kubota S, Sasaki Y, Redpath P, Migaud ME, Apte RS, Uchida K et al (2016) Long‐term administration of nicotinamide mononucleotide mitigates age‐associated physiological decline in mice. Cell Metab 24: 795–806
    1. Mitchell SJ, Bernier M, Aon MA, Cortassa S, Kim EY, Fang EF, Palacios HH, Ali A, Navas‐Enamorado I, Di Francesco A et al (2018) Nicotinamide improves aspects of healthspan, but not lifespan mice. Cell Metab 27: 667–676
    1. Morris BJ (2013) Seven sirtuins for seven deadly diseases of aging. Free Radic Biol Med 56: 133–171
    1. Mouchiroud L, Houtkooper RH, Moullan N, Katsyuba E, Ryu D, Canto C, Mottis A, Jo YS, Viswanathan M, Schoonjans K et al (2013) The NAD(+)/sirtuin pathway modulates longevity through activation of mitochondrial UPR and FOXO signaling. Cell 154: 430–441
    1. Muraki K, Han L, Miller D, Murnane JP (2013) The role of ATM in the deficiency in nonhomologous end‐joining near telomeres in a human cancer cell line. PLoS Genet 9: e1003386
    1. O'Callaghan N, Baack N, Sharif R, Fenech M (2012) A qPCR‐based assay to quantify oxidized guanine and other FPG‐sensitive base lesions within telomeric DNA. Biotechniques 51: 403–411
    1. Partida‐Sanchez S, Cockayne DA, Monard S, Jacobson EL, Oppenheimer N, Garvy B, Kusser K, Goodrich S, Howard M, Harmsen A et al (2001) Cyclic ADP‐ribose production by CD38 regulates intracellular calcium release, extracellular calcium influx and chemotaxis in neutrophils and is required for bacterial clearance in vivo . Nat Med 7: 1209–1216
    1. de Picciotto NE, Gano LB, Johnson LC, Martens CR, Sindler AL, Mills KF, Imai S, Seals DR (2016) Nicotinamide mononucleotide supplementation reverses vascular dysfunction and oxidative stress with aging in mice. Aging Cell 15: 522–530
    1. Sahin E, Colla S, Liesa M, Moslehi J, Muller FL, Guo M, Cooper M, Kotton D, Fabian AJ, Walkey C et al (2011) Telomere dysfunction induces metabolic and mitochondrial compromise. Nature 470: 359–365
    1. Sarkar J, Liu Y (2016) The origin of oxidized guanine resolves the puzzle of oxidation‐induced telomere‐length alterations. Nat Struct Mol Biol 23: 1070–1071
    1. Savage SA (2018) Beginning at the ends: telomeres and human disease. F1000Research 7: 524–538
    1. Scheibye‐Knudsen M, Ramamoorthy M, Sykora P, Maynard S, Lin PC, Minor RK, Wilson DM 3rd, Cooper M, Spencer R, de Cabo R et al (2012) Cockayne syndrome group B protein prevents the accumulation of damaged mitochondria by promoting mitochondrial autophagy. J Exp Med 209: 855–869
    1. Schultz MB, Sinclair DA (2016) Why NAD(+) declines during aging: it's destroyed. Cell Metab 23: 965–966
    1. Tang BL (2016) Sirt1 and the mitochondria. Mol Cells 39: 87–95
    1. Tarrago MG, Chini CCS, Kanamori KS, Warner GM, Caride A, de Oliveira GC, Rud M, Samani A, Hein KZ, Huang R et al (2018) A potent and specific CD38 inhibitor ameliorates age‐related metabolic dysfunction by reversing tissue NAD(+) decline. Cell Metab 27: 1081–1095
    1. Vallabhaneni H, Zhou F, Maul RW, Sarkar J, Yin J, Lei M, Harrington L, Gearhart PJ, Liu Y (2015) Defective repair of uracil causes telomere defects in mouse hematopoietic cells. J Biol Chem 290: 5502–5511
    1. Vannini N, Campos V, Girotra M, Trachsel V, Rojas‐Sutterlin S, Tratwal J, Ragusa S, Stefanidis E, Ryu D, Rainer PY et al (2019) The NAD‐booster nicotinamide riboside potently stimulates hematopoiesis through increased mitochondrial clearance. Cell Stem Cell 24: 405–418
    1. Verdin E (2015) NAD(+) in aging, metabolism, and neurodegeneration. Science 350: 1208–1213
    1. Wan B, Yin J, Horvath K, Sarkar J, Chen Y, Wu J, Wan K, Lu J, Gu P, Yu EY et al (2013) SLX4 assembles a telomere maintenance toolkit by bridging multiple endonucleases with telomeres. Cell Rep 4: 861–869
    1. Yang SJ, Choi JM, Kim L, Park SE, Rhee EJ, Lee WY, Oh KW, Park SW, Park CY (2014) Nicotinamide improves glucose metabolism and affects the hepatic NAD‐sirtuin pathway in a rodent model of obesity and type 2 diabetes. J Nutr Biochem 25: 66–72
    1. Yang Y, Sauve AA (2016) NAD(+) metabolism: bioenergetics, signaling and manipulation for therapy. Biochim Biophys Acta 1864: 1787–1800
    1. Yap TA, Plummer R, Azad NS, Helleday T (2019) The DNA damaging revolution: PARP inhibitors and beyond. Am Soc Clin Oncol Educ Book 39: 185–195
    1. Yoshino J, Baur JA, Imai SI (2018) NAD(+) intermediates: the biology and therapeutic potential of NMN and NR. Cell Metab 27: 513–528
    1. Zhang H, Ryu D, Wu Y, Gariani K, Wang X, Luan P, D'Amico D, Ropelle ER, Lutolf MP, Aebersold R et al (2016) NAD(+) repletion improves mitochondrial and stem cell function and enhances life span in mice. Science 352: 1436–1443
    1. Zhu XH, Lu M, Lee BY, Ugurbil K, Chen W (2015) In vivo NAD assay reveals the intracellular NAD contents and redox state in healthy human brain and their age dependences. Proc Natl Acad Sci USA 112: 2876–2881

Source: PubMed

3
Abonner