pharmacological bypass of nad+ salvage pathway protects ... · pharmacological bypass of nad+...

6
Pharmacological bypass of NAD + salvage pathway protects neurons from chemotherapy- induced degeneration Hui-wen Liu a , Chadwick B. Smith a , Mark S. Schmidt b , Xiaolu A. Cambronne a,1 , Michael S. Cohen c , Marie E. Migaud d , Charles Brenner b,2 , and Richard H. Goodman a,2 a Vollum Institute, Oregon Health & Science University, Portland, OR 97239; b Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 55242; c Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, OR 97239; and d Mitchell Cancer Institute, University of South Alabama, Mobile, AL 33604 Contributed by Richard H. Goodman, August 22, 2018 (sent for review June 4, 2018; reviewed by Michael P. Coleman and William Lee Kraus) Axon degeneration, a hallmark of chemotherapy-induced periph- eral neuropathy (CIPN), is thought to be caused by a loss of the essential metabolite nicotinamide adenine dinucleotide (NAD + ) via the prodegenerative protein SARM1. Some studies challenge this notion, however, and suggest that an aberrant increase in a direct precursor of NAD + , nicotinamide mononucleotide (NMN), rather than loss of NAD + , is responsible. In support of this idea, blocking NMN accumulation in neurons by expressing a bacterial NMN dea- midase protected axons from degeneration. We hypothesized that protection could similarly be achieved by reducing NMN produc- tion pharmacologically. To achieve this, we took advantage of an alternative pathway for NAD + generation that goes through the intermediate nicotinic acid mononucleotide (NAMN), rather than NMN. We discovered that nicotinic acid riboside (NAR), a precursor of NAMN, administered in combination with FK866, an inhibitor of the enzyme nicotinamide phosphoribosyltransferase that produces NMN, protected dorsal root ganglion (DRG) axons against vincristine- induced degeneration as well as NMN deamidase. Introducing a dif- ferent bacterial enzyme that converts NAMN to NMN reversed this protection. Collectively, our data indicate that maintaining NAD + is not sufficient to protect DRG neurons from vincristine- induced axon degeneration, and elevating NMN, by itself, is not sufficient to cause degeneration. Nonetheless, the combination of FK866 and NAR, which bypasses NMN formation, may provide a therapeutic strategy for neuroprotection. chemotherapy-induced peripheral neuropathy | axon degeneration | NAD + | nicotinamide mononucleotide | nicotinic acid riboside A xon degeneration is an early feature of chemotherapy- induced peripheral neuropathy (CIPN), a severe and often permanent condition caused by anticancer agents that perturb microtubule depolymerization, such as taxanes (paclitaxel), vinca alkaloids (vincristine, vinblastine), and proteasome inhibitors (bortezomib) (1). The asymmetrical shape of peripheral neurons, with axons that can extend for meters, renders these cells par- ticularly sensitive to insults that impair fast axonal transport. The numbness, pain, and heat/cold hyperalgesia associated with CIPN can be extremely troubling to patients, potentially limiting the use of otherwise effective chemotherapeutic agents (2). In many ways, CIPN is reminiscent of Wallerian degeneration, a self-destructive process involving the distal portion of trans- ected axons characterized by a beaded appearance, retraction, and axonal disintegration (3). Initially attributed to a failure to transport essential nutrients, Wallerian degeneration is now thought to result from an intrinsic axon destruction pathway. A key finding in this more recent understanding was the discovery of a spontaneously occurring mouse mutation, Wallerian degeneration slow (Wld s ), which prevented degeneration of transected axons (4). Molecular characterization of Wld s revealed a fusion protein containing the nicotinamide adenine dinucleotide (NAD + ) biosyn- thetic enzyme nicotinamide mononucleotide adenylyltransferase 1 (NMNAT1) linked to a sequence that mislocalizes this normally nuclear protein to the cytoplasm (5, 6). The rescue engendered by Wld s is consistent with the observation that NMNAT2, the pre- dominant cytoplasmic isoform of the enzyme, becomes depleted in axons after transection because of its short half-life (7). This de- pletion was predicted to result in decreased levels of axonal NAD + (8), an idea supported by the findings that protection from de- generation could be achieved by providing the NAD + precursors, nicotinamide mononucleotide (NMN) or nicotinamide riboside (NR), augmenting activity of nicotinamide phosphoribosyl- transferase (NAMPT) (9), or increasing NMNAT expression (3) (Fig. 1A). A second key observation was the finding that an intrinsic axon degeneration pathway depended on the prodegenerative protein, SARM1 (10). Initially identified in Drosophila, the SARM1 ortholog in mammalian neurons is both necessary and sufficient for axonal degeneration (11). Moreover, CIPN, as well as other models of Wallerian degeneration such as posttraumatic brain injury, can be rescued by the loss of SARM1 (12). The finding that SARM1 contains an intrinsic NAD + hydrolase activity sup- ported the idea that NAD + plays a central role in maintaining Significance Axon degeneration is caused by multiple chemotherapeutic agents and is a hallmark of chemotherapy-induced peripheral neuropathy (CIPN). CIPN is associated with the loss of the es- sential metabolite nicotinamide adenine dinucleotide (NAD + ), a vital metabolite involved in energy homeostasis, but whether NAD + loss or the accumulation of an NAD + precursor, nicotin- amide mononucleotide (NMN), is responsible for axon degen- eration is controversial. We found that axon degeneration caused by vincristine, a widely used chemotherapeutic agent, could be ameliorated by forcing peripheral neurons to use an NAD + biosynthetic pathway that bypasses NMN formation. This strategy provides an approach for preventing CIPN. Author contributions: H.-w.L., C.B., and R.H.G. designed research; H.-w.L., C.B.S., and M.S.S. performed research; X.A.C., M.S.C., and M.E.M. contributed new reagents/analytic tools; H.-w.L., M.S.S., and R.H.G. analyzed data; and H.-w.L., C.B., and R.H.G. wrote the paper. Reviewers: M.P.C., Babraham Institute and Cambridge Centre for Brain Repair; and W.L.K., The University of Texas Southwestern Medical Center at Dallas. Conflict of interest statement: C.B. owns stock in and consults for ChromaDex, Inc., and consults for Cytokinetics, Inc. M.E.M. has consulted for ChromaDex, Inc. Published under the PNAS license. 1 Present address: Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712. 2 To whom correspondence may be addressed. Email: [email protected] or [email protected]. This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. 1073/pnas.1809392115/-/DCSupplemental. Published online September 26, 2018. 1065410659 | PNAS | October 16, 2018 | vol. 115 | no. 42 www.pnas.org/cgi/doi/10.1073/pnas.1809392115 Downloaded by guest on October 19, 2020

Upload: others

Post on 04-Aug-2020

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Pharmacological bypass of NAD+ salvage pathway protects ... · Pharmacological bypass of NAD+ salvage pathway protects neurons from chemotherapy-induced degeneration Hui-wen Liu a,

Pharmacological bypass of NAD+ salvage pathwayprotects neurons from chemotherapy-induced degenerationHui-wen Liua, Chadwick B. Smitha, Mark S. Schmidtb, Xiaolu A. Cambronnea,1, Michael S. Cohenc, Marie E. Migaudd,Charles Brennerb,2, and Richard H. Goodmana,2

aVollum Institute, Oregon Health & Science University, Portland, OR 97239; bDepartment of Biochemistry, Carver College of Medicine, University of Iowa,Iowa City, IA 55242; cDepartment of Physiology and Pharmacology, Oregon Health & Science University, Portland, OR 97239; and dMitchell Cancer Institute,University of South Alabama, Mobile, AL 33604

Contributed by Richard H. Goodman, August 22, 2018 (sent for review June 4, 2018; reviewed by Michael P. Coleman and William Lee Kraus)

Axon degeneration, a hallmark of chemotherapy-induced periph-eral neuropathy (CIPN), is thought to be caused by a loss of theessential metabolite nicotinamide adenine dinucleotide (NAD+) viathe prodegenerative protein SARM1. Some studies challenge thisnotion, however, and suggest that an aberrant increase in a directprecursor of NAD+, nicotinamide mononucleotide (NMN), ratherthan loss of NAD+, is responsible. In support of this idea, blockingNMN accumulation in neurons by expressing a bacterial NMN dea-midase protected axons from degeneration. We hypothesized thatprotection could similarly be achieved by reducing NMN produc-tion pharmacologically. To achieve this, we took advantage of analternative pathway for NAD+ generation that goes through theintermediate nicotinic acid mononucleotide (NAMN), rather thanNMN. We discovered that nicotinic acid riboside (NAR), a precursorof NAMN, administered in combination with FK866, an inhibitor ofthe enzyme nicotinamide phosphoribosyltransferase that producesNMN, protected dorsal root ganglion (DRG) axons against vincristine-induced degeneration as well as NMN deamidase. Introducing a dif-ferent bacterial enzyme that converts NAMN to NMN reversedthis protection. Collectively, our data indicate that maintainingNAD+ is not sufficient to protect DRG neurons from vincristine-induced axon degeneration, and elevating NMN, by itself, is notsufficient to cause degeneration. Nonetheless, the combinationof FK866 and NAR, which bypasses NMN formation, may providea therapeutic strategy for neuroprotection.

chemotherapy-induced peripheral neuropathy | axon degeneration |NAD+ | nicotinamide mononucleotide | nicotinic acid riboside

Axon degeneration is an early feature of chemotherapy-induced peripheral neuropathy (CIPN), a severe and often

permanent condition caused by anticancer agents that perturbmicrotubule depolymerization, such as taxanes (paclitaxel), vincaalkaloids (vincristine, vinblastine), and proteasome inhibitors(bortezomib) (1). The asymmetrical shape of peripheral neurons,with axons that can extend for meters, renders these cells par-ticularly sensitive to insults that impair fast axonal transport. Thenumbness, pain, and heat/cold hyperalgesia associated withCIPN can be extremely troubling to patients, potentially limitingthe use of otherwise effective chemotherapeutic agents (2).In many ways, CIPN is reminiscent of Wallerian degeneration,

a self-destructive process involving the distal portion of trans-ected axons characterized by a beaded appearance, retraction,and axonal disintegration (3). Initially attributed to a failure totransport essential nutrients, Wallerian degeneration is nowthought to result from an intrinsic axon destruction pathway. Akey finding in this more recent understanding was the discovery ofa spontaneously occurring mouse mutation, Wallerian degenerationslow (Wlds), which prevented degeneration of transected axons(4). Molecular characterization of Wlds revealed a fusion proteincontaining the nicotinamide adenine dinucleotide (NAD+) biosyn-thetic enzyme nicotinamide mononucleotide adenylyltransferase 1

(NMNAT1) linked to a sequence that mislocalizes this normallynuclear protein to the cytoplasm (5, 6). The rescue engendered byWlds is consistent with the observation that NMNAT2, the pre-dominant cytoplasmic isoform of the enzyme, becomes depleted inaxons after transection because of its short half-life (7). This de-pletion was predicted to result in decreased levels of axonal NAD+

(8), an idea supported by the findings that protection from de-generation could be achieved by providing the NAD+ precursors,nicotinamide mononucleotide (NMN) or nicotinamide riboside(NR), augmenting activity of nicotinamide phosphoribosyl-transferase (NAMPT) (9), or increasing NMNAT expression(3) (Fig. 1A).A second key observation was the finding that an intrinsic axon

degeneration pathway depended on the prodegenerative protein,SARM1 (10). Initially identified in Drosophila, the SARM1ortholog in mammalian neurons is both necessary and sufficientfor axonal degeneration (11). Moreover, CIPN, as well as othermodels of Wallerian degeneration such as posttraumatic braininjury, can be rescued by the loss of SARM1 (12). The finding thatSARM1 contains an intrinsic NAD+ hydrolase activity sup-ported the idea that NAD+ plays a central role in maintaining

Significance

Axon degeneration is caused by multiple chemotherapeuticagents and is a hallmark of chemotherapy-induced peripheralneuropathy (CIPN). CIPN is associated with the loss of the es-sential metabolite nicotinamide adenine dinucleotide (NAD+), avital metabolite involved in energy homeostasis, but whetherNAD+ loss or the accumulation of an NAD+ precursor, nicotin-amide mononucleotide (NMN), is responsible for axon degen-eration is controversial. We found that axon degenerationcaused by vincristine, a widely used chemotherapeutic agent,could be ameliorated by forcing peripheral neurons to use anNAD+ biosynthetic pathway that bypasses NMN formation.This strategy provides an approach for preventing CIPN.

Author contributions: H.-w.L., C.B., and R.H.G. designed research; H.-w.L., C.B.S., andM.S.S. performed research; X.A.C., M.S.C., and M.E.M. contributed new reagents/analytictools; H.-w.L., M.S.S., and R.H.G. analyzed data; and H.-w.L., C.B., and R.H.G. wrotethe paper.

Reviewers: M.P.C., Babraham Institute and Cambridge Centre for Brain Repair; andW.L.K., The University of Texas Southwestern Medical Center at Dallas.

Conflict of interest statement: C.B. owns stock in and consults for ChromaDex, Inc., andconsults for Cytokinetics, Inc. M.E.M. has consulted for ChromaDex, Inc.

Published under the PNAS license.1Present address: Department of Molecular Biosciences, University of Texas at Austin,Austin, TX 78712.

2To whom correspondence may be addressed. Email: [email protected] [email protected].

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1809392115/-/DCSupplemental.

Published online September 26, 2018.

10654–10659 | PNAS | October 16, 2018 | vol. 115 | no. 42 www.pnas.org/cgi/doi/10.1073/pnas.1809392115

Dow

nloa

ded

by g

uest

on

Oct

ober

19,

202

0

Page 2: Pharmacological bypass of NAD+ salvage pathway protects ... · Pharmacological bypass of NAD+ salvage pathway protects neurons from chemotherapy-induced degeneration Hui-wen Liu a,

axonal integrity after injury (13). Whether NAD+ loss and theassociated defects in energy metabolism are the primary effec-tors of axonal degeneration is controversial, however. NAD+

hydrolysis mediated by in vitro translated SARM1 generatesother bioactive molecules, namely, ADP ribose (ADPR) andcyclic ADP ribose (cADPR) (14), both of which can cause po-tentially deleterious elevations in intracellular Ca2+ (15).Although there is considerable evidence supporting the idea

that loss of NAD+ contributes to axon degeneration, otherstudies suggest a different model; namely, that axonal injury andthe subsequent loss of NMNAT2 activity increases levels of theNMNAT2 substrate NMN, and that it is the elevated level of

NMN that triggers degeneration. Support for this idea includesthe findings that a degree of protection was afforded by FK866,an NAMPT inhibitor, and that reducing NMN levels in neuronsby expressing Escherichia coli NMN deamidase (NMNd), a bac-terial enzyme that converts NMN to its deamidated form, nic-otinic acid mononucleotide (NAMN), protected axons as well asWlds or eliminating SARM1 (16, 17). Reconciling these two modelshas been difficult because elevating NMN levels by increasingNAMPT expression has also been shown to be protective (9).Although inducing expression of genes such as Wlds and NMNd

were effective in preventing Wallerian degeneration, these ap-proaches have limited potential for clinical translation. We soughtto take advantage of the well-characterized pathways for NAD+

synthesis to develop a pharmacological approach for CIPNtreatment, aiming to identify a small molecule or combination ofsmall molecules that would eliminate NMN accumulation whilemaintaining NAD+ levels. The three classical pathways for NAD+

biosynthesis use tryptophan (the de novo pathway), nicotinic acid(NA; the Preiss-Handler pathway), and nicotinamide (NAM; theNAM salvage pathway). The de novo and Preiss-Handler path-ways converge at the level of NAMN, whereas the NAM salvagepathway and the recently identified NR kinase pathway (18) gothrough NMN.We hypothesized, therefore, that use of the formertwo pathways, which do not produce NMN, might be beneficial forpreventing CIPN. As a model for CIPN, we examined axon de-generation in dorsal root ganglion (DRG) neurons treated withthe chemotherapeutic agent, vincristine. We found that the denovo and Preiss-Handler pathways are not functional in DRGs,and that these neurons relied on the NAM salvage pathway forNAD+ synthesis. To reroute the pathway for NAD+ synthesis toone that avoided NMN, we employed a deamidated form of NR,nicotinic acid riboside (NAR) (19). Similar to NR, NAR is con-verted to its product, in this instance NAMN, through the actionsof the widely expressed NR kinases, NRK1 and NRK2 (18, 20). Ofnote, nerve injury has been shown to induce expression of theNRK gene products (9, 20), potentially increasing the efficacy ofNAR in supporting NAD+ synthesis.Although NAR treatment delayed the axon degeneration

caused by vincristine, it was only marginally more effective thanNR and did not reduce NMN levels below those in DRGs treatedwith vincristine alone. To decrease NMN production further, weblocked NAMPT activity with the inhibitor FK866. This combi-nation of NAR and FK866 reduced NMN levels and protected theneurons as well as NMNd. In addition, we showed that expressingthe Francisella tularensis NMN synthetase (NMNsyn), an enzymethat converts NAMN to NMN, reversed this protection. The ef-fects of FK866 and NAR administered in combination provide therationale for a therapeutic strategy for preventing CIPN.

ResultsNAR Can Maintain NAD+ Synthesis in DRG Neurons. The principalroutes for NAD+ synthesis in mammalian cells are depicted inFig. 1A. To determine which NAD+ biosynthetic pathways arefunctional in DRG neurons, we performed RT-qPCR analysis ofmRNA isolated from rat E15 DRGs maintained for 6 d in culture(DIV6) and treated with deoxyfluorouridine to minimize glialcontamination. We found that NA phosphoribosyltransferases(NAPRT) and quinolinic acid phosphoribosyltransferases were\expressed at low levels compared with NAMPT, with NRK1 andNAD+ synthetase at intermediate levels (Fig. 1B). Treatment ofcells with NA, NR, or FK866 did not change the expression pat-tern (SI Appendix, Fig. S1A). These results suggested that mostNAD+ production in DRG neurons occurs through the NAMsalvage pathway. Moreover, because of the relatively low expres-sion of NAPRT, we predicted that NA would not be able to serveas an effective NAD+ precursor. In contrast, NAR, which re-quires only NRK to produce NAMN, should be able to produceNAD+ in the absence of NAPRT. To test this hypothesis, we

A

B C

D

Fig. 1. NAR maintains NAD+ synthesis in DRG neurons that lack NAPRT. (A)Diagram of NAD+ synthesis pathways. The de novo pathway converts tryp-tophan (Trp) to quinolinic acid (QA) and then, via quinolinic acid phos-phoribosyltransferase (QPRT), to NAMN. NAMN is converted to NAAD and,ultimately, NAD+, via NMNAT and NAD+ synthetase (NADS). NAMN can alsobe produced from nicotinic acid and NAR via NAPRT and NRK, respectively.NAM and NR are converted to NMN by NAMPT and NRK1 and NRK2, asindicated, and NMNAT converts NMN to NAD+. FK866 is a specific inhibitorof NAMPT. NMNd and NMNsyn are bacterial enzymes that convert NMN toNAMN and NAMN to NMN, respectively. Axon injury activates SARM1, whichhydrolyzes NAD+ to produce ADPR, cADPR, and NAM. (B) Relative levels ofNAD+ biosynthetic enzymes in DRG neurons determined by RT-qPCR(mean ± SD; n = 3; P < 0.005 for each gene compared with NAMPT). (C)Levels of NAD+ in DRG neurons 16 h after treatment with 100 nM FK866 and500 μM nicotinic acid or NAR (mean ± SD; n = 3; **P < 0.005). (D) Repre-sentative images (Left) and quantitation of tetramethylrhodamine methylester staining (Right) 24 h after exposure to the indicated agents. F0 repre-sents the signal intensity after addition of the uncoupling agent FCCP.

Liu et al. PNAS | October 16, 2018 | vol. 115 | no. 42 | 10655

BIOCH

EMISTR

Y

Dow

nloa

ded

by g

uest

on

Oct

ober

19,

202

0

Page 3: Pharmacological bypass of NAD+ salvage pathway protects ... · Pharmacological bypass of NAD+ salvage pathway protects neurons from chemotherapy-induced degeneration Hui-wen Liu a,

used quantitative targeted NAD+ metabolomics (21) to ascertainwhether NA or NAR could support NAD+ levels in DRG neu-rons treated with the NAMPT inhibitor, FK866. As expected,NAD+ levels fell in DRGs treated with FK866 and were notrestored by NA (Fig. 1C). In contrast, NAR restored NAD+ levelsto normal even when NAMPT was inhibited. To test whether thispharmacological rerouting of NAD+ synthesis pathways couldsustain mitochondrial function, we examined mitochondrial mem-brane potential using tetramethylrhodamine methyl ester. Nodiminution in tetramethylrhodamine methyl ester staining wasevident in DRGs treated with NAR and FK866 (Fig. 1D), andaxon integrity was maintained (SI Appendix, Fig. S1B). Takentogether, these experiments indicate that although NAD+ syn-thesis in DRG neurons normally occurs via NAM salvage, NAD+

production can be redirected to a deamidated pathway by com-bining NAR and FK866, bypassing the putatively neurotoxic NMNintermediate.

NR and NAR Delay Vincristine-Induced Axon Degeneration. Chemo-therapeutic drugs are thought to mediate axon degeneration byactivating SARM1, leading to a loss of NAD+ and production ofcADPR and ADPR (Fig. 1). Many of these drugs, particularlythose that interfere with microtubule polymerization and de-polymerization, reduce levels of axonal NMNAT2, which ispredicted to increase NMN accumulation in addition to contrib-uting to the depression of NAD+. We confirmed these changesby measuring NAD+ and NMN levels in E15 DRG neuronstreated with 50 nM vincristine at DIV6. After 16 h of vincristinetreatment, NMN levels increased from undetectable to 14.5 ±6.8 pmol/106 cells and NAD+ fell to about 40% of control levels(Fig. 2A). The observation that the alternative NR kinase sub-strate, NAR, produces NAMN rather than NMN (19) led us totest whether NAR might be more effective than NR in protectingaxons from the effects of vincristine. Further, we thought theseagents might help sort out whether the NMN increase or NAD+

decrease is responsible for degeneration. For these experiments,we treated DRG neurons with 500 μM NR or NAR at the sametime that vincristine was administered and assayed for NMN andNAD+ 16 h later. As expected, NMN levels in DRGs treated withvincristine were significantly higher after NR than NAR, whereasboth treatments restored NAD+ to the basal level. It was not

possible to follow NMN or NAD+ levels at later times becausethe axons displayed significant degeneration.To examine how these treatments affected axon degeneration,

we treated cells with NR or NAR at the time of vincristine ex-posure and monitored axon degeneration at various times aftertreatment, shown in representative images 72 h after treatment(Fig. 2B) and in determination of the degeneration index (DI;Fig. 2C and SI Appendix, SI Materials and Methods). Both treat-ments delayed degeneration compared with vincristine alone, butNAR was only slightly more effective than NR despite the dif-ference in NMN accumulation. By 96 h, no protection was evi-dent after either agent. Taken together, these studies showedthat restoration of NAD+ levels afforded by NR and NAR onlypartially protected against axon degeneration. Consistent with theidea that NMN appears to be a result of NMNAT2 depletion (17),NAR treatment did not reduce the vincristine-induced accu-mulation of NMN, possibly accounting for the limited degree ofprotection. In addition, consistent with the reversibility of theNMNAT reaction (22), some NMN accumulation was also seenin DRGs treated with NAR and FK866 in the absence of vin-cristine (SI Appendix, Fig. S2).

Combining NAR with FK866 Prevents Axon Degeneration. Previousstudies showed that E. coli NMNd protected primary neuroncultures from Wallerian degeneration, presumably by convertingNMN to NAMN (17). This protection was reminiscent of thatprovided by Wlds, which has been shown to lower NMN levels ininjured axons (16). In an effort to recapitulate the actions ofNMNd and Wlds pharmacologically, we tested whether reroutingNAD+ synthesis from the NAM salvage pathway to the deami-dated pathway could protect axons from degeneration as well asthese genetic approaches. We first examined effects of FK866,which blocks formation of NMN from NAM. FK866 eliminatedvincristine-induced NMN production, but at the expense ofdrastically reducing levels of NAD+ (Fig. 3A). To augmentNAD+ production, we added 500 μM NAR to the regimen. In-terestingly, NAD+ levels after treatment with vincristine, FK866,and NAR were approximately the same as after vincristine alone,and still substantially lower than levels in untreated neurons.However, this combination significantly depressed NMN accu-mulation compared with vincristine alone (note difference inscale between Figs. 2 and 3). For comparison, we showed that a

A B C

Fig. 2. NR and NAR delay but do not prevent vincristine-induced axon degeneration. (A) Levels of NMN (Top) and NAD+ (Bottom) in DRG neurons treatedwith 50 nM vincristine. At the same time as vincristine on DIV6, 500 μM NR or NAR were added, and cells were collected for LC-MS 16 h later. (B) Repre-sentative images of DRG axons 72 h after vincristine treatment. NAR or NR were added at same time as vincristine. (C) DI (Methods) at various times aftertreatment. For A, statistics were determined using one-way ANOVA. ns, not significant; *P < 0.05; **P < 0.005; ***P < 0.0005; ****P < 10−4 denote differencesusing Bonferroni’s multiple comparisons test (mean ± SD; n ≥ 3 for each treatment). For C, statistics were determined using two-way ANOVA; asterisks denoteP values between indicated treatments, using Bonferroni’s multiple comparisons test (n = 12 for each time).

10656 | www.pnas.org/cgi/doi/10.1073/pnas.1809392115 Liu et al.

Dow

nloa

ded

by g

uest

on

Oct

ober

19,

202

0

Page 4: Pharmacological bypass of NAD+ salvage pathway protects ... · Pharmacological bypass of NAD+ salvage pathway protects neurons from chemotherapy-induced degeneration Hui-wen Liu a,

lentivirus expressing E. coli NMNd completely eliminated NMNaccumulation.We then tested the combination of NAR and FK866 in axonal

protection assays, treating DRG neurons with both agents at thetime of vincristine administration. As shown in representativeimages (Fig. 3B) and DI assays (Fig. 3C), the combination of NARand FK866 protected neurons as well as NMNd expression. Takentogether, these experiments indicate that substantial protectionfrom vincristine-induced axon degeneration can be achieved bysimultaneously blocking NMN accumulation and providing analternative source for NAD+. The FK866 and NAR combinationdid not increase NAD+ levels back to normal, however, suggestingthat the key determinant for axon protection is reducing NMNaccumulation. Thus, a depressed level of NAD+, when combinedwith low levels of NMN, is not sufficient for axon degeneration.

F. tularensis NMNsyn Reverses Protection Afforded by NAR and FK866.To test the idea that the combination of NAR plus FK866 pro-tected against axon degeneration by depressing NMN levels, weinfected DRGs with a lentivirus expressing NMNsyn. This en-zyme functions in a manner opposite to NMNd, convertingNAMN to NMN. In vincristine-treated neurons, the depressionin NMN accumulation achieved by treatment with FK866 andNAR (compare lanes 1 and 2 of Fig. 4A) was reversed by ex-pression of NMNsyn, and NAD+ levels were restored (Fig. 4A,lane 3). NMNsyn, when combined with NAR, increased NMN tothe level detected in DRG neurons treated with vincristine andelevated NAD+ to levels above normal. In degeneration assays ofDRGs treated with vincristine, NMNsyn largely reversed theprotective effect of NAR plus FK866 (Fig. 4 B and D), whereasNMNsyn administered with NAR did not cause degeneration

A B C

Fig. 3. Combination of FK866 and NAR protects axons from degeneration. (A) NMN (Top) and NAD+ (Bottom) levels in DRGs exposed to 50 nM vincristineand treated with the indicated compounds or transduced at DIV2 with a lentivirus expressing NMNd. NAR and FK866 were added at the same time asvincristine on DIV6, and cells were collected for LC-MS analysis 16 h later (mean ± SD; n ≥ 3 for each treatment). (B) Representative images of DRG axons at72 h posttreatment. Control represents neurons treated with vincristine alone. (C) Quantitation of axon degeneration at various times after vincristineadministration. Asterisks denote P values as earlier (n = 12 for each time).

A CB

D

Fig. 4. NMNsyn reverses protection afforded by FK866 plus NAR. (A) NMN (Top), and NAD+ (Bottom) levels in DRGs transduced with a lentivirus expressing F.tularensis NMNsyn or control on DIV2. Cells were cultured for 5 d after infection and treated with 50 nM vincristine, FK866, and NAR, as indicated (mean ± SD;n ≥ 3 for each treatment). (B) Representative images of DRG neurons at 72 h posttreatment with the agents indicated. (C) Representative images of NMNSyn-expressing DRG neurons at 72 h post-NAR treatment. (Axons were not treated with vincristine.) (D) Quantitation of axon degeneration. Asterisks denote Pvalues as earlier (n = 12 for each time).

Liu et al. PNAS | October 16, 2018 | vol. 115 | no. 42 | 10657

BIOCH

EMISTR

Y

Dow

nloa

ded

by g

uest

on

Oct

ober

19,

202

0

Page 5: Pharmacological bypass of NAD+ salvage pathway protects ... · Pharmacological bypass of NAD+ salvage pathway protects neurons from chemotherapy-induced degeneration Hui-wen Liu a,

when vincristine was absent (Fig. 4C). Taken together, theseexperiments indicate that shunting NAD+ synthesis from apathway through NAMN to one that involves NMN eliminatesthe protection from vincristine-induced degeneration. In the ab-sence of vincristine, however, the rise in NMN was insufficient toinduce degeneration if NAD+ levels were elevated. Consistent witha previous study (23), NR alone similarly increased NMN levels,elevated NAD+, and did not cause degeneration (SI Appendix, Fig.S3). In contrast, blocking the salvage pathway via FK866 treatmentcaused degeneration after 72 h (SI Appendix, Fig. S3B).The idea behind using NMNd was to reduce levels of the

potentially neurotoxic NMN, but this manipulation should alsoincrease flux through the deamidated NAD+ synthesis pathway(Fig. 1A). NAR uses this pathway as well, and should similarlyincrease levels of deamidated NAD+ precursors. It is possiblethat some of these deamidated intermediates could contribute tothe axon protective effects of NMNd or the FK866 plus NARcombination by blocking SARM1 activity. To test whether fluxthrough the deamidated pathway was augmented, we quantifiedlevels of nicotinic acid adenine dinucleotide (NAAD), the finalstep in the deamidated pathway before NAD+ production (24).As anticipated, levels of NAAD were increased in DRGs treatedwith vincristine and NAR, NMNd, or the FK866 plus NARcombination (SI Appendix, Fig. S4). NMNsyn eliminated theNAAD accumulation mediated by FK866 plus NAR.SARM1 contains an amino-terminal autoinhibitory domain

that prevents its activity under basal conditions. Removal of thisinhibitory domain reveals an enzymatic activity that allows thehydrolysis of NAD+ to produce ADPR and cADPR (14). To testwhether a deamidated NAD+ precursor such as NAAD mightinterfere with SARM1 activity, we purified epitope-tagged SARM1,either full-length or the active SARM1 fragment, from HEK293cells and tested its ADPR cyclase activity in vitro. No interferencewith NAD+ hydrolysis or cADPR generation was detected (SI Ap-pendix, Fig. S5) in the presence of NAAD, suggesting that NAADdoes not function as a SARM1 inhibitor.

DiscussionWallerian degeneration has served as a model of multiple neu-rological conditions, but perhaps its most common manifestationis in CIPN, a frequent adverse effect of many commonly usedchemotherapeutic agents. Insights into the mechanisms under-lying Wallerian degeneration provide the hope of developingagents capable of preventing or reversing this serious condition.Genetic studies in Drosophila and mouse models led to the dis-covery of two key determinants of Wallerian degeneration;namely, Wlds (4) and SARM1 (10). The finding that loss ofSARM1 phenocopiesWlds suggested that the two might intersecton a common pathway, and this idea was supported by the ob-servations that Wlds contains an NAD+ biosynthetic enzyme andSARM1 has an NAD+ hydrolase activity. Regardless of whetherSARM1 promotes axon degeneration by depleting NAD+ or bygenerating ADPR and cADPR, pharmacologic agents capable ofinhibiting SARM1 would be good candidates for CIPN pre-vention. Unfortunately, no such agents have been developed.Our approach was to address the consequences of Wlds andSARM1 action; namely, effects on the NAD+ metabolome.There is general agreement that axon injury leads to a loss of

NMNAT2 and a decrease in NAD+ (3). How this relates to axondegeneration is controversial, however. Although considerableevidence supports the idea that NAD+ loss, and perhaps thegeneration of bioactive NAD+ metabolites, are major factors (9,13), some studies have suggested that build-up of the NMNAT2substrate NMN is more important (16, 17). Obviously, these twomodels lead to opposing predictions; in the first, it should bebeneficial to increase NAD+ production by providing NAD+

precursors, whereas in the second, efforts should be devoted todepressing elevated NMN levels. In this report, we attempt to

reconcile the two models and provide a pharmacologic approachto preventing CIPN. We call this approach NAR addiction be-cause it renders DRG neurons dependent on an exogenousprecursor not normally used in NAD+ synthesis that feeds intothe normally vestigial deamidated pathway.Consistent with the second model, using quantitative targeted

metabolomics, we observed a persistent elevation of NMN levelsafter vincristine treatment. Unlike a previous study that showedthe elevated NMN levels decreasing to baseline at 6 h afteraxotomy (23), we found that the levels remained elevated for 24h. This discrepancy could be a result of different types of injury,axotomy versus vincristine, improved internal standards, or thatwe quantify whole-cell NMN levels as opposed to those in axonsalone. NR, commonly used to prevent conditions associated withNAD+ loss (20, 25, 26), protected against the vincristine-induceddecrease in NAD+, but generated even higher levels of NMN. Inan effort to reduce NMN formation pharmacologically, wetreated DRGs with NAR, a precursor of NAMN that feeds intothe deamidated pathway (Fig. 1). The Preiss-Handler pathwaynormally depends on NAPRT, the NA-dependent equivalent ofNAMPT. NAPRT mRNA levels in DRG neurons were found tobe extremely low, however, rendering this pathway nonfunctional.NAR, similar to NR, requires only phosphorylation by NRK1/NRK2 to generate the next intermediate in the NAD+ biosynthesispathway. Both precursors restored NAD+ levels to normal, butNAR caused a substantially smaller increase in NMN. Still, NARdid not eliminate the vincristine-induced increase in NMN, whichmight account for the residual propensity for degeneration. Wesuggest that this NMN is generated from the activity of NAMPT,which produces NMN from nicotinamide, and from the bidirec-tional activity of NMNAT. Notably, our results indicated that re-storing NAD+ levels to normal was not sufficient for full protection.Consistent with observations of others (17), we found that E.

coli NMNd greatly depressed NMN levels in neurons treatedwith vincristine and protected neurons from degeneration. Inan effort to reduce NMN levels pharmacologically, we used acombination of agents that simultaneously blocked the NAMsalvage pathway while supporting NAD+ production through thedeamidated pathway. In the presence of vincristine, FK866greatly depressed NAD+ levels, as expected from the block oftwo salvage pathway enzymes, and this decrease was partiallyrescued by adding NAR. The combination of FK866 and NARwas as protective as E. coli NMNd in axon degeneration assays,however. NAD+ levels in neurons treated with vincristine, NAR,and FK866 were no greater than those observed after vincristinealone. This suggests that the loss of NAD+ does not fully accountfor vincristine-induced axon degeneration.As a further test of the idea that NAR plus FK866 protected

axons by eliminating production of NMN, we examined the ef-fects of F. tularensis NMNsyn, whose activity is the opposite ofthe E. coli NMNd. NMNsyn shunts the Preiss-Handler in-termediate NAMN to NMN, allowing NAR to feed into thesalvage pathway at the level of NMN. NMNsyn severely limitedthe axonal protection afforded by FK866 and NAR, apparentlyby reestablishing neurotoxic levels of NMN. Because NAD+

levels in neurons expressing NMNsyn returned to baseline, thesestudies suggest that the NMN increase, rather than NAD+ loss,was responsible for degeneration. One caveat of our studies isthat in addition to depressing NMN levels, NAR increased levelsof the deaminated NAD+ precursor, NAAD. However, we foundthat NAAD did not reduce cADPR generation by purified, full-length SARM1 or a SARM1 fragment lacking the autoinhibitorydomain, suggesting that NAAD inhibition of SARM1 activityis unlikely.It has been suggested that SARM1 inhibitors might be useful

for axon protection, but to date, no SARM1 inhibitors have beendeveloped. The agents we have used target pathways known tobe affected by SARM1 and are readily available. It could be

10658 | www.pnas.org/cgi/doi/10.1073/pnas.1809392115 Liu et al.

Dow

nloa

ded

by g

uest

on

Oct

ober

19,

202

0

Page 6: Pharmacological bypass of NAD+ salvage pathway protects ... · Pharmacological bypass of NAD+ salvage pathway protects neurons from chemotherapy-induced degeneration Hui-wen Liu a,

imagined that elevating cellular NAD+ content might promotegrowth of transformed cells, but the combination of NAR withFK866 did not lead to supraphysiological NAD+ levels, andneurons treated with this combination appeared normal (SIAppendix, Fig. S1) and maintained normal mitochondrial iongradients.Taken together, our results largely support the idea that NMN

promotes axon degeneration. Depressing levels of NMN confersaxon protection even in the face of lower NAD+ levels. However,elevating NMN was not sufficient to cause axon degeneration, asneurons treated with NAR and NMNsyn displayed an elevationin NMN but did not degenerate. It is possible that the aug-mented level of NAD+ in this setting was protective (17). Inaddition, how NMN relates mechanistically to axon degenera-tion, and presumably SARM1 activation, remains unresolved. Thereis no evidence supporting the notion that NMN regulates SARM1directly, raising the possibility that some other NMN-binding pro-tein is required in SARM1 regulation.Finally, the viability of NAR-addicted neurons has implica-

tions for how mitochondrial NAD+ levels are maintained. Inthese neurons, there is no cytoplasmic NMN, and all NAD+ is

derived through the deamidated pathway. The final step in thispathway depends on NAD+ synthetase, an enzyme that is ex-cluded from mitochondria (27). Thus, all NAD+ in the NMN-depleted cells must be produced in the cytoplasm, and as aconsequence, mitochondrial NAD+ must be transported fromthe cytoplasm rather than generated in the matrix via NMNAT3.This finding supports our observation that knockdown of NMNAT2in other cell types reduced mitochondrial NAD+ levels (28). Mi-tochondrial NAD+ transporters have been identified in yeast andbacteria, but have not been identified in mammals.

Materials and MethodsAnimal use followed NIH guidelines and complied with Oregon Health andScience University Institutional Animal Care and Use Committee (IACUC)under an approved protocol. Methods of cell culture, lentiviral packaging,NAD+ metabolite extraction, LC-MS/MS analysis, axon degeneration as-say, and statistical analyses are provided in SI Appendix, SI Materialsand Methods.

ACKNOWLEDGMENTS. This work was supported by NIH Grants 1R01AG055431-01 (to R.H.G.) and DP2GM126897 (to X.A.C.), Roy J. Carver Trust (C.B.), and PewBiomedical Scholar (M.S.C.).

1. Grisold W, Cavaletti G, Windebank AJ (2012) Peripheral neuropathies from chemo-therapeutics and targeted agents: Diagnosis, treatment, and prevention. Neuro-oncol14:iv45–iv54.

2. Park SB, et al. (2013) Chemotherapy-induced peripheral neurotoxicity: A criticalanalysis. CA Cancer J Clin 63:419–437.

3. Conforti L, Gilley J, Coleman MP (2014) Wallerian degeneration: An emerging axondeath pathway linking injury and disease. Nat Rev Neurosci 15:394–409.

4. Lunn ER, Perry VH, Brown MC, Rosen H, Gordon S (1989) Absence of Wallerian de-generation does not hinder regeneration in peripheral nerve. Eur J Neurosci 1:27–33.

5. Coleman MP, et al. (1998) An 85-kb tandem triplication in the slow Wallerian de-generation (Wlds) mouse. Proc Natl Acad Sci USA 95:9985–9990.

6. Mack TG, et al. (2001) Wallerian degeneration of injured axons and synapses is de-layed by a Ube4b/Nmnat chimeric gene. Nat Neurosci 4:1199–1206.

7. Gilley J, Coleman MP (2010) Endogenous Nmnat2 is an essential survival factor formaintenance of healthy axons. PLoS Biol 8:e1000300.

8. Wang J, et al. (2005) A local mechanism mediates NAD-dependent protection of axondegeneration. J Cell Biol 170:349–355.

9. Sasaki Y, Araki T, Milbrandt J (2006) Stimulation of nicotinamide adenine dinucleo-tide biosynthetic pathways delays axonal degeneration after axotomy. J Neurosci26:8484–8491.

10. Osterloh JM, et al. (2012) dSarm/Sarm1 is required for activation of an injury-inducedaxon death pathway. Science 337:481–484.

11. Gerdts J, Summers DW, Sasaki Y, DiAntonio A, Milbrandt J (2013) Sarm1-mediated axondegeneration requires both SAM and TIR interactions. J Neurosci 33:13569–13580.

12. Henninger N, et al. (2016) Attenuated traumatic axonal injury and improved func-tional outcome after traumatic brain injury in mice lacking Sarm1. Brain 139:1094–1105.

13. Gerdts J, Brace EJ, Sasaki Y, DiAntonio A, Milbrandt J (2015) SARM1 activation trig-gers axon degeneration locally via NAD+ destruction. Science 348:453–457.

14. Essuman K, et al. (2017) The SARM1 Toll/Interleukin-1 receptor domain possessesintrinsic NAD+ cleavage activity that promotes pathological axonal degeneration.Neuron 93:1334–1343.

15. Cantó C, Menzies KJ, Auwerx J (2015) NAD(+) metabolism and the control of energyhomeostasis: A balancing act between mitochondria and the nucleus. Cell Metab 22:31–53.

16. Di Stefano M, et al. (2015) A rise in NAD precursor nicotinamide mononucleotide(NMN) after injury promotes axon degeneration. Cell Death Differ 22:731–742.

17. Di Stefano M, et al. (2017) NMN deamidase delays Wallerian degeneration and res-cues axonal defects caused by NMNAT2 deficiency in vivo. Curr Biol 27:784–794.

18. Bieganowski P, Brenner C (2004) Discoveries of nicotinamide riboside as a nutrientand conserved NRK genes establish a Preiss-Handler independent route to NAD+ infungi and humans. Cell 117:495–502.

19. Tempel W, et al. (2007) Nicotinamide riboside kinase structures reveal new pathwaysto NAD+. PLoS Biol 5:e263.

20. Vaur P, et al. (2017) Nicotinamide riboside, a form of vitamin B3, protects againstexcitotoxicity-induced axonal degeneration. FASEB J 31:5440–5452.

21. Trammell SA, Brenner C (2013) Targeted, LCMS-based metabolomics for quantitativemeasurement of NAD(+) metabolites. Comput Struct Biotechnol J 4:e201301012.

22. Kornberg A (1950) Reversible enzymatic synthesis of diphosphopyridine nucleotideand inorganic pyrophosphate. J Biol Chem 182:779–793.

23. Sasaki Y, Nakagawa T, Mao X, DiAntonio A, Milbrandt J (2016) NMNAT1 inhibits axondegeneration via blockade of SARM1-mediated NAD+ depletion. eLife 5:e19749.

24. Bieganowski P, Pace HC, Brenner C (2003) Eukaryotic NAD+ synthetase Qns1 containsan essential, obligate intramolecular thiol glutamine amidotransferase domain re-lated to nitrilase. J Biol Chem 278:33049–33055.

25. Trammell SA, et al. (2016) Nicotinamide riboside opposes type 2 diabetes and neu-ropathy in mice. Sci Rep 6:26933.

26. Diguet N, et al. (2018) Nicotinamide riboside preserves cardiac function in a mousemodel of dilated cardiomyopathy. Circulation 137:2256–2273.

27. Nikiforov A, Dölle C, Niere M, Ziegler M (2011) Pathways and subcellular compart-mentation of NAD biosynthesis in human cells: From entry of extracellular precursorsto mitochondrial NAD generation. J Biol Chem 286:21767–21778.

28. Cambronne XA, et al. (2016) Biosensor reveals multiple sources for mitochondrialNAD+. Science 352:1474–1477.

Liu et al. PNAS | October 16, 2018 | vol. 115 | no. 42 | 10659

BIOCH

EMISTR

Y

Dow

nloa

ded

by g

uest

on

Oct

ober

19,

202

0