来自:jlc108 > 馆藏分类
配色: 字号:
辛克莱:NAD+前体将成为对抗新冠等病毒感染的重要武器
2023-01-18 | 阅:  转:  |  分享 
  
storms (see Glossary). A key to effective interventions is to trigger robust antiviral defenses

but with checked in?ammation. Therefore, molecules that suppress both viral replication and

Aging

Highlights

NAD

+

can exert both antiviral and anti-

in?ammatoryeffectsinmiceandhumans,

which might be bene?cial during co-

ronavirus disease 2019 (COVID-19)

infections.

Compared with healthy individuals,

NAD

+

concentrations in tissues and or-

gansarelowerinolderindividualsandin

patients with diseases associated with

severe COVID-19 symptoms, including

diabetes and cardiovascular disease.

Viral infections, including coronavirus

infections, have been reported to further

deplete cellular NAD

+

stores.

Many NAD

+

-dependent enzymes,

including members of the sirtuin and

poly-ADP-ribose polymerase (PARP)

families, display potent antiviral activities.

Severe acute respiratory syndrome co-

ronavirus 2 (SARS-CoV-2) and a variety

of viruses possess ADP-ribosyl hydrolase

activity, which counteracts the activity of

PARPs.

Viruses such as SARS-CoV-2 can

hyperactivate the immune system

by activating nuclear factor kappa B

(NF-κB) and the NOD-, LRR-, and

pyrin domain-containing protein 3

(NLRP3) in?ammasome, potentially

leading to deadly cytokine storms.

NAD

+

-boostingcompoundsmaycoun-

teract these processes.

Several NAD

+

-boosting compounds

and molecules that target NAD

+

-

producing or -consuming enzymes

are in clinical development as putative

anti-in?ammatory or antiviral drugs.

Institute, Paul F. Glenn Center for

Trends in

more susceptible to developing severe disease could lead to new candidate strategies for

therapeutic intervention.

Biology of Aging Research, Harvard

Medical School, Boston, MA, USA

2

These authors contributed equally.

diseases af?ict boththe very oldand young,makingthisdemographicpatternfor COVID-19 sus-

ceptibility unusual [5]. A better understanding of the mechanisms that render older individuals

AtragicandpoorlyunderstoodaspecttoCOVID-19istheincreasedsusceptibilityoftheelderlyto

severe forms of the disease [3]. Rates of hospitalization, intensive care unit (ICU) admission, and

death increase with age, while the young are often left relatively unscathed [4]. Most infectious

1

Department of Genetics, Blavatnik

in?ammation may be particularly important for ?ghting severe COVID-19 [1]. A growing body

of evidence shows that the metabolite NAD

+

is a mediator of both antiviral and anti-

in?ammatory mechanisms. Based on this evidence, we posit that therapies that boost

NAD

+

concentrations might play a role in preventing and treating severe COVID-19 and

other viral infections.

Firstdescribedasa yeastfermentationfactoroveracentury ago,today, NAD

+

hasrisentoprom-

inence as a regulator of healthy aging. Low levels of NAD

+

in tissues and organs are associated

with aging, metabolic syndrome, and in?ammation, while dietary interventions that slow age-

related diseases increase NAD

+

concentrations [2]. Here, we review the epidemiological and

mechanistic data supporting a role for NAD

+

in modulating the outcomes of viral infections,

with a focus on SARS-CoV and SARS-CoV-2. We also explore ongoing approaches to boost

NAD

+

levels for therapeutic bene?t in the clinic.

NAD

+

and risk factors for severe COVID-19

Opinion

NAD

+

in COVID-19 and viral infections

Minyan Zheng ,

1,2

Michael B. Schultz,

1,2

and David A. Sinclair

1,



NAD

+

, as an emerging regulator of immune responses during viral infections, may

be a promising therapeutic target for coronavirus disease 2019 (COVID-19). In this

Opinion, we suggest that interventions that boost NAD

+

levels might promote anti-

viral defenseand suppress uncontrolled in?ammation. Wediscuss the association

between low NAD

+

concentrations and risk factors for poor COVID-19 outcomes,

including aging and common comorbidities. Mechanistically, we outline how viral

infectionscanfurtherdepleteNAD

+

anditsrolesinantiviraldefenseandin?amma-

tion. We also describe how coronaviruses can subvert NAD

+

-mediated actions via

genes that remove NAD

+

modi?cations and activate the NOD-, LRR-, and pyrin

domain-containing protein 3 (NLRP3) in?ammasome. Finally, we explore ongoing

approaches to boost NAD

+

concentrations in the clinic to putatively increase

antiviral responses while curtailing hyperin?ammation.

NAD

+

as a modulator of viral infection outcomes

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and other viral infections

render the body a battle?eld, requiring mobilization of vast resources to mount an effective

defense. These defenses can back?re when uncontrolled, resulting in deadly cytokine

Immunology

Trends in Immunology, April 2022, Vol. 43, No. 4 https://doi.org/10.1016/j.it.2022.02.001 283

? 2022 Elsevier Ltd. All rights reserved.

Correspondence:

david_sinclair@hms.harvard.edu

(D.A. Sinclair).

Trends in Immunology

Wepositthatoneculpritmaybeade?ciency in NAD

+

, the concentration of which decreases

during aging in nearly every tissue studied across species [6]. In rodents, this decrease has been

observed in tissues and cells that are relevant to COVID-19 infection, such as the skeletal muscle,

liver endothelial cells, and macrophages [7–10]. A growing body of human data corroborates an

association between age and low NAD

+

concentrations across multiple tissues including the

skin, blood, liver, and muscle [10–13,117].

WhyNAD

+

levelsfallduringagingisanactiveareaofresearch.Studiesinmicehaveimplicatedan

increase in the concentrations and activity of NAD

+

-consuming enzymes such as CD38, poly

(ADP-ribose) polymerase 1 (PARP1)1, and sterile alpha and TIR motif containing 1 (SARM1)

[9,14–17], as well as an insuf?cient ?ux from NAD

+

-producing enzymes such as nicotinamide

phosphoribosyltransferase (NAMPT), indoleamine 2,3-dioxygenase (IDO), and quinolinate

phosphoribosyltransferase (QPRT) [10,18–20]. These enzymes all serve as potential pharmaco-

logical targets to raise NAD

+

concentrations (Figure 1).

Comorbidities

NAD

+

concentrations are also low in certain comorbidities associated with COVID-19

severity. One such comorbidity is insulin resistance (IR) and diabetes mellitus [21]. Speci?cally, in

mouse models, genetic (KK/HIJ), diet-induced (high-fat diet), streptozotocin-induced, and

age-associated IR have been associated with lower NAD

+

concentrations in liver and white adipose

tissue, while restoration of NAD

+

with NAD

+

boosters such as nicotinamide ribose (NR) and

nicotinamide mononucleotide (NMN) (oral, intraperitoneal,or infusion) reverses IR[22–26]. In humans,

metabolic syndrome and obesity have been associated with low NAD

+

concentrations in adipose

tissue [27]. NMN was recently shown to improve insulin sensitivity in prediabetic women, and further

clinical studies are ongoing, discussed later in this review [28](Table 1, Key table). In addition, a

recently developed oral antidiabetic medication, imeglimin, can enhance glucose-stimulated ATP

generation and induce the synthesis of NAD

+

in pancreatic islets derived from diseased rodents

with type 2 diabetes [29].

Another risk factor for severe COVID-19 is cardiovascular disease (CVD) [30]. Several preclinical

studies demonstrate that raising NAD

+

concentrations in endothelial cells reverses vascular and

endothelialdysfunction[8,31].Furtherstudieshaveshownabene?cialroleforNAD

+

incardiovas-

cular function in mouse models, including models of dyslipidemia, ischemia–reperfusion injury,

and diastolic heart failure [32,33]. This link is supported in humans by epidemiological data

demonstrating a correlation between dietary intake of the NAD

+

precursor niacin (nicotinic acid)

and vascular health, including brachial-artery ?ow-mediated dilation and serum low-density lipo-

protein (LDL) concentrations [34]. In fact, niacin is an USA FDA-approved therapy for reducing

LDL, ApoB, and triglycerides, and for raising high-density lipoprotein (HDL). Additionally, multiple

ongoing clinical studies are testing the effects of other NAD

+

boosters such as NMN and NR on

hypertension and heart failure (discussed later) (Table 1).

Mechanisms of NAD

+

in viral infections

Viral infections can deplete NAD

+

concentrations

Not only are low NAD

+

concentrations associated with risk factors for poor COVID-19 outcomes,

butcertainviralinfectionscanfurtherdepleteNAD

+

ininfectedcells.Forexample,lowerNAD

+

con-

centrations have been reported in human peripheral blood leukocytes infected with HIV-1 in vitro

[35],human ?broblastsinfectedwithherpessimplexvirus1(HSV-1)[36],andintheskeletalmuscle

of individuals coinfected with HIV-1 and hepatitis C virus [37]. These effects are presumably due to

the induction of NAD

+

-consuming enzymes such as CD38 and PARPs [17]. For instance, CD8

+

T lymphocytes expressing CD38 have been proposed as a marker for HIV-1-mediated disease

284 Trends in Immunology, April 2022, Vol. 43, No. 4

Trends in Immunology

progression [38]. The decline of NAD

+

concentrations in human ?broblasts induced by HSV-1

infection is associated with increased protein poly(ADP-ribosyl)ation, and can be blocked by phar-

macological inhibition of PARP1/PARP2 [36].

Trendsrends inin ImmunologyImmunology

Figure 1. NAD

+

metabolism and points of pharmacological intervention. Enzymes involved in NAD

+

biosynthesis

and hydrolysis play important roles in in?ammation and immunity. Biosynthetic pathways include the NAD

+

salvage

pathway, which recycles nicotinamide to form NMN, then NAD

+

, and the de novo pathway that begins with tryptophan.

Hydrolysis of NAD

+

is largely carried out by PARPs, which tag target proteins with poly- or mono-(ADP ribose); sirtuins,

which remove acyl groups and create O-acyl-ADP-ribose; and CD38, BST, and SARM1, which create (cyclic)-ADP-ribose

[44]. There are multiple points of potential pharmacological intervention throughout NAD

+

metabolic pathways. Created

with BioRender.com. Abbreviations: i, inhibitor; NAMPT, nicotinamide phosphoribosyltransferase; NMN, nicotinamide

mononucleotide; NR, nicotinamide ribose; NSP3, nonstructural protein 3; PARP, poly(ADP-ribose) polymerase; SARM1,

sterile alpha and TIR motif containing 1; SIRT, sirtuin.

Glossary

Apoptosis-associated speck-like

protein containing a caspase

recruitment domain (ASC):

in?ammasome adaptor that plays a key

role inthe assemblyand activationofthe

in?ammasome. The NLRP3

in?ammasome consists of a sensor

(NLRP3), an adaptor (ASC), and an

effector (caspase-1). Upon

sensing-speci?c triggers, NLRP3

undergoes conformational changes

which catalyze ASC oligomerization to

form a macromolecular signaling complex

known as the ASC speck. ASC then

recruits caspase-1. Active caspase-1

cleaves pro-IL-1β and pro-IL-18, which

are vital cytokines during infection and

in?ammation.

Cytokine storm: life-threatening event

triggered by uncontrolled in?ammatory

responses with the release of a large

amounts of proin?ammatory cytokines,

including IL-6, IL-1, TNFα, and IFN. This

leads to the recruitment and expansion

of various immune cells into injury sites

which can result in tissue and organ

damage.

Histone H3 lysine 9 (H3K9)

deacetylation:epigeneticmodi?cation;

H3K9 is generally acetylated when a

gene is transcriptionally activated and is

deacetylated (and/or methylated) when

ageneissilenced.

Imeglimin: novel oral agent for the

treatment of type 2 diabetes. Its

mechanism of action involves

ampli?cation of glucose-stimulated insulin

secretion and enhanced insulin action.

Approved for use in Japan in 2021.

P2X purinoceptor 7: expressed in an

increasingnumberofcelltypes;primarily

mediates in?ammation and cell death;

ligand-gatedcationchannelactivated by

high concentrations of extracellular ATP,

triggering the assembly and activation of

the NLRP3 in?ammasome and

subsequent release of IL-1β and IL-18.

Poly(ADP-ribose) polymerases

(PARPs): with coenzyme NAD

+

,PARP

enzymes catalyze ADP-ribosylation or the

transferofADP-ribosylgroupsfrom NAD

+

to nucleophilic side chains of proteins.

They can also ADP-ribosylate DNA and

RNA. In humans, there are 17 identi?ed

PARPs. PARPs participate in diverse

cellular functions such as DNA repair,

apoptosis, unfolded protein response,

pathogen response, and in?ammation.

Sirtuins: enzymes that couple NAD

+

degradation to deacylation (e.g.,

deacetylation, desuccinylation, and

Trends in Immunology, April 2022, Vol. 43, No. 4 285

demyristoylation). With NAD

+

, sirtuins

remove acyl groups from lysine residues

on proteins, sense intracellular NAD

+

concentrations, and transduce a signal

via protein deacylation. In humans, there

are seven sirtuins at different cellular

locations that regulate diverse functions,

including transcription, genome stability,

metabolism, and cell signaling.

Stress granules (SGs): dense,

membraneless, organelles in the cytosol

that appear in response to cellular stress

to promotecellsurvival.Ribonucleoprotein

assemblies that store mRNAs stalled at

translation initiation. The introduction of

viral RNA into the cytoplasm triggers the

formation of stress granules.

TNF receptor-associated factor 3

(TRAF3): member of the TRAF family;

contains a RING domain with E3

ubiquitin ligase activity, and a TRAF

domain mediating protein–protein

interactions; functions in in?ammation,

antiviral defense, and apoptosis. There

are seven TRAF proteins identi?ed in

mammals.

Toll/interleukin-1 receptor domain:

intracellular signaling domain in proteins

that mediates protein–protein

interactions between Toll-like receptors

and signal transduction proteins. When

activated, the TIR domain recruits

cellular adaptor proteins and induces

downstream activation of kinases.

Trends in Immunology

Similar depletion of NAD

+

has been reported with coronavirus infections. Speci?cally, blood from

severe COVID-19 patients contains lower amounts of the NAD

+

precursor NMN compared with

blood from healthy individuals [39]. Moreover, expression changes in genes involved in NAD

+

synthesis and utilization such as Nampt, Parp9, Parp12, and Parp14 have been observed in ep-

ithelial cell lines and enterocyte organoids infected withSARS-CoV-2 compared withmock infec-

tion[40].SimilarNAD

+

-relatedgeneexpressionchangeswerealsoreportedinthelungtissueofa

deceased COVID-19 patient and in the bronchoalveolar lavage ?uid of SARS-CoV-2-infected in-

dividuals relative to healthy controls [40]. Furthermore, infection of a coronavirus, mouse hepatitis

virus (MHV), in mouse bone-marrow-derived macrophages (BMDMs) induced NAD

+

depletion

[40] as well as increased gene expression of many PARPs including Parp7, Parp9, Parp10,

Parp11,Parp12,Parp13,andParp14relativetomockinfection[41].Theseobservationssuggest

that NAD

+

metabolic pathways are under increased demand during SARS-CoV-2 infection, and

highlights the potential relevance of NAD

+

in modulating COVID-19 disease outcomes.

NAD

+

-consuming enzymes in antiviral mechanisms

NAD

+

harbors an important role in fueling the activity of enzymes that regulate mammalian

immune responses [42,43]. Classically, NAD

+

participates in redox processes, but it also

participates in non‐redox reactions in which it is hydrolyzed, and it non‐enzymatically regulates

protein–protein interactions [16](Figure 1). In these latter functions, NAD

+

acts as a signaling

molecule, serving as a marker of energy availability and directing a cell to respond to metabolic

changes via the action of NAD

+

-utilizing enzymes [44].

PARPs and sirtuins are two NAD

+

-dependent enzyme families that participate in immune

responses [42,43]. By adding or removing post-translational modi?cations on key proteins

such as nuclear factor kappa B (NF-κB), they can coordinate the intensity of in?ammatory and

immune responses [42]. This places NAD

+

in an important position for both promoting strong

immune responses to pathogens, and for keeping those responses in check.

PARP1, the pre-eminent member of the PARP family, is a potent coactivator of the proin?amma-

tory transcription factor NF-κB, and therefore participates in initiating speci?c immune responses

[42]. However, this is a double-edged sword, as PARP1 may also increase the severity of cyto-

kine storms as it regulates the expression of many NF-κB-dependent cytokines and chemokines

[42]. Indeed, inhibiting or deleting PARP1 has ameliorated the severity of symptoms in several

in?ammatory disease rodent models including asthma and colitis [45–51]. For example, PARP1

inhibition (pharmacological or genetic) has prevented ovalbumin-induced lung in?ammation in

mice [45] and in a guinea pig model of asthma [46]. Treatment with PARP inhibitors has attenu-

ated in?ammation associated with colitis seenininterleukin-10(IL-10)de?cient (Il10

-/-

)mice

[47], as well as in trinitrobenzene sulfonic acid-treated rats [49]. Furthermore, PARP1 knockout

(KO) (Parp1

-/-

) mice are protected from dextran sulfate sodium-induced colitis compared with

wild-type mice [48].

Several PARPs harbor potent antiviral functions [42,52–59]. Indeed, PARP13 is a powerful anti-

viralfactor thatrecognizesvariousviruses fromseveralfamilies,including Retroviridae, Filoviridae,

Alphaviridae, and Hepadnaviridae [53–56]. It binds to speci?c sequences of viral RNAs during in-

fection and mediates their degradation via the cellular mRNA decay machinery; however, these

functions are not dependent on PARP-mediated ADP-ribosylation [54–56]. Expression of

PARP7, PARP10, and the long isoform of PARP12 (PARP12L) ef?ciently inhibits cellular transla-

tion and the replication of Venezuelan equine encephalitis virus and other alphaviruses in verte-

brate cells [52,59]. These effects of PARP12L are dependent on its catalytic activity. Moreover,

PARP9 and PARP14 are also upregulated in macrophages stimulated by interferon (IFN)-γ and

286 Trends in Immunology, April 2022, Vol. 43, No. 4

Key table

Table 1. Selected clinical trials involving NAD

+

boosters

Clinicaltrials.

gov ID

Phase Interventions Duration Type Enrollment

(participants)

Inclusion criteria Primary endpoint Completion

NCT03151239 N/A NMN, 250 mg/d or

placebo

8 wk Randomized,

double-blind,

placebo-controlled

25 Prediabetic

postmenopausal

women age 55–75 yr

Change in muscle

insulin sensitivity

June 2021

NCT05175768 N/A NMN, NMN

+L-leucine, or

placebo

Up to 28

d

Randomized,

double-blind,

placebo-controlled

375 Individuals age >40 yr

hospitalized with

COVID-19 requiring

supplemental oxygen

COVID-19

associated fatigue

December

2022

NCT04903210 IV NMN, 800 mg/d +

lifestyle modi?cation

or lifestyle

modi?cation alone

8 wk Randomized, single

blind

20 Individuals age 18–65

yr with mild essential

hypertension (BP

130/80–159/99)

Hypertension

(?ow-mediated

dilation and

brachial–ankle pulse

wave velocity)

July 2022

NCT04664361 N/A NMN 250 mg/d,

NMN 500 mg/d, or

placebo

38 d Randomized,

double-blind,

placebo-controlled

150 Healthy men age

20–49 yr with regular

moderate physical

activity

Muscle recovery

(post-endurance

Wingate Anaerobic

Test)

September

2022

NCT02950441 II NR 1 g/d or

placebo

21 d,

followed

by

washout

and

crossover

Randomized,

double-blind,

placebo-controlled,

crossover

12 Men age 70–80 yr Mitochondrial

function

(respirometry) and

NAD

+

concentrations

in muscle biopsy

September

2019

NCT02921659 I/II NR 1 g/d or

placebo

6wk,

followed

by

crossover

Randomized,

double-blind,

placebo-controlled,

crossover

30 Individuals age 55–79

yr

Treatment-emergent

adverse events

October

2016

NCT04040959 II NR 1 g/d or

placebo

3 mo Randomized,

double-blind,

placebo-controlled

118 Individuals age 35–80

yr with chronic kidney

disease stage III or IV

Arterial stiffness

(carotid–femoral

pulse wave velocity)

September

2024

NCT03821623 II NR 1 g/d or

placebo

3 mo Randomized,

double-blind,

placebo-controlled

118 Individuals age ≥50 yr

with systolic blood

pressure between

120 and 139 mmHg

Resting systolic

blood pressure

December

2023

NCT04528004 I NR dose escalation

to 1 g/d or placebo

14 d Randomized,

double-blind,

placebo-controlled

40 Adults with end-stage

heart failure NYHA

class IV

Whole blood NAD

+

concentrations

August

2024

NCT04407390 II NR 1 g/d or

placebo

14 d Randomized,

double-blind,

placebo-controlled

100 Individuals age ≥70 yr

with COVID-19

Hypoxic respiratory

failure

May 2022

NCT04818216 II NR 1 g/d or

placebo

10 d Randomized,

double-blind,

placebo-controlled

100 Adults hospitalized

with COVID-19 and

acute kidney injury

Whole blood NAD

+

concentrations,

adverse events,

thrombocytopenia

June 2023

NCT04573153 II/III NR + serine +

L-carnitine tartrate +

N-acetylcysteine +

hydroxychloroquine

vs. placebo +

hydroxychloroquine

14 d Randomized,

placebo-controlled

400 Adults with

COVID-19,

ambulatory and

symptomatic

Hospitalization rate March

2021

(continued on next page)

Trends in Immunology

Trends in Immunology, April 2022, Vol. 43, No. 4 287

Primary endpoint Completion

Cognitive functioning

measured by

executive functioning

and memory

composite scores

December

2022

Trends in Immunology

haveopposing rolesinmacrophageactivation[58].PARP9 activatesIFNγ–STAT1 signaling and

induces proin?ammatory activation while PARP14 ADP-ribosylation reduces STAT1 phos-

phorylationinIFNγ-treatedhumanmacrophages[58].,Additionally,thenucleocapsidproteins

of several coronaviruses, including SARS-CoV, MERS-CoV, and MHV, are ADP-ribosylated in

infected cells, presumably by PARPs, which may indicate a common use of this pathway

among viruses. However, the functional consequences of such ADP‐ribosylation remain to

be investigated [57]. We argue that since PARP enzymatic activity requires NAD

+

[44], main-

taining a suf?cient NAD

+

concentration may be crucial for achieving PARP-related antiviral

mechanisms.

Sirtuins also play a role in antiviral defenses [43,60–64]. Indeed, sirtuin 1 (SIRT1) KO or inhibition

promotes the lifecycle and replication of vesicular stomatitis virus in mouse embryonic ?broblasts

(MEFs) and Kaposi’s sarcoma-associated herpesvirus in human lymphoma cell lines [61,62].

Disruption of SIRT1 also increases HPV16 E1–E2 replication [60]. Moreover, knockdown via

siRNA of each of the seven sirtuins in human ?broblast cells promoted the growth of a di-

verse set of human viruses after infection, including human cytomegalovirus (CMV), HSV1,

adenovirus type 5, and in?uenza virus (H1N1) [63]. Furthermore, SIRT1-activating drugs

such as resveratrol and CAY10602 have inhibited the replication of these viruses [63].

SIRT6 promotes tumor necrosis factor (TNF)α secretion, as evidenced from the suppression

of TNFα release from SIRT6 KO MEFs, whereby TNFα secretion would be expected to pro-

mote the eradication of pathogens [64]. Indeed, pharmacological inhibition and siRNA

knockdown of SIRT6 and NAMPT, an NAD

+

-synthetic enzyme (Figure 1), in mouse ?bro-

Table 1. (continued)

Clinicaltrials.

gov ID

Phase Interventions Duration Type Enrollment

(participants)

Inclusion criteria

NCT04809974 IV NR 2 g/d or

placebo

22 wk Randomized,

double-blind,

placebo-controlled

100 Individuals age 18–65

yr, 2+ mo out from

COVID-19 PCR

diagnosis, currently

PCR negative, with

persistent cognitive

and physical

dif?culties

(long-COVID)

blasts promoted CMV replication [65].

Other NAD

+

-utilizing enzymes that also play roles during immune responses include CD38

[66], BST1 [2], and SARM1 [43]. Indeed, CD38 and BST1 are highly expressed on the sur-

face of macrophages and lymphocytes and produce extracellular cyclic(ADP-ribose), a

calcium-mobilizing second messenger that is important for immune cell activation [2,66,67].

SARM1, another potent NADase, contains a Toll/IL-1 receptor domain, which might elicit

neuroprotective innate immune responses, as suggested from mouse models of neurode-

generation [68].

Viruses ?ghting back

One antiviral mechanism that is associated with PARP activity is the formation of cytosolic,

membraneless organelles called stress granules (SGs) to sequester viral RNAs and arrest pro-

viraltranslation[69,70].Indeed,ADP-ribosylationofSGcomponentsbyPARPspromotesthefor-

mation of antiviral SGs [71–73]. Several antiviral PARPs including PARP5a, 12, 13, 14, and 15

localize to SGs in human cells, presumably to execute this modi?cation [71].

288 Trends in Immunology, April 2022, Vol. 43, No. 4

Trends in Immunology

As a counter-mechanism to the host, many viruses, including coronaviruses and alphaviruses,

deploy ADP-ribosylhydrolases to remove SG-promoting ADP-ribose modi?cations [74–77].

SARS-CoV-2 has such a domain with mono-ADP-ribosylhydrolase activity in its largest

encoded protein, nonstructural protein 3 (NSP3) [79,80]. Ectopic expression of the SARS-

CoV-2NSP3macrodomainreversesPARP9-dependent ADP-ribosylationoftargetproteins

[81]. In live viruses, this domain is important for both viral replication and virulence. Mutations

of this domain in MHV and SARS-CoV led to attenuated viral replication, and thus, the mutant

viruses were unable to cause lung disease in infected mice, while treatment with pan-PARP in-

hibitors enhanced SARS-CoV replication and inhibited IFN production in BMDMs infected

with macrodomain-de?cient mutant coronavirus [41,82]. These ?ndings suggest that the

SARS-CoV-2 NSP3 protein might also interfere with SG formation and thereby allow for

evasion of cellular antiviral responses (Figure 2). We argue that providing more NAD

+

to

fuel the activity of PARPs might shift the antiviral balance back in favor of host cells.

Viruses can drive uncontrolled in?ammation

One of the potential catastrophic effects of SARS-CoV-2 infections is cytokine storms. Indeed,

the concentrations of circulating proin?ammatory factors such as IL-1, IL-6, and TNFα are

strongly associated with ICU admission and mortality in COVID-19 patients [83,84]. Studies of

the ?rst SARS pandemic coronavirus, SARS-CoV, have shown that viral proteins play an active

role in the processing and release of the two speci?cproin?ammatory cytokines, IL-1β and

IL-18, by enhancing NF-κB transcriptional activity and promoting the formation of the NLRP3

in?ammasome (Figure 2)[85–90].

The SARS-CoV proteins that facilitate these processes include open reading frame (ORF)3a,

envelope protein (E), and ORF8b [85–90]. ORF3a mediates NF-κB activation through

TRAF3-dependent ubiquitination of an NF-κB inhibitory subunit, and mediates speck formation

of the in?ammasome subunit ASC, which accompanies assembly of the NLRP3 in?ammasome

in human cells [86]. Additionally, ORF3a has transmembrane domains and ion channel (IC)

activity that drives K

+

ef?ux, which further promotes activation of the NLRP3 in?ammasome.

Indeed, IL-1β secretion was completely blocked when BMDMs, stimulated with lentiviruses

expressing the SARS-CoV ORF3a, were treated with K

+

-rich medium [78,85]. The SARS-CoV E

protein also promotes in?ammasome activation through its intracellular activity; it forms lipid–protein

channels at the endoplasmic reticulum (ER)–Golgi intermediate compartment, and the resulting Ca

2

+

stimulates the activation of the NLRP3 in?ammasome. The E protein also promotes IL-1β release

in mammalian cells expressing the NLRP3 in?ammasome, while E protein mutants lacking ion

conductance cannot boost IL-1β secretion [87,88]. ORF8b causes in?ammasome activation in

human macrophages and interacts directly with NLRP3 in vitro; moreover, it can form protein

aggregates and trigger ER stress and lysosomal damage that activate the in?ammasome in HeLa

cells [89]. These processes not only promote virulence but may also facilitate viral replication

[87,90]. For example, a comparative study of the functional motifs included within the SARS-CoV

viroporins showed that full-length E and ORF3a proteins were required for maximal SARS-CoV

replication and virulence in infected mice [87].

As with SARS-CoV, SARS-CoV-2 can also activate the in?ammasome [91]. Active NLRP3

in?ammasomes have been found in peripheral blood mononuclear cells (PBMCs) and tissues

from deceased COVID-19 patients upon autopsy, along with higher concentrations of serum

IL-18 which correlated with COVID-19 severity. SARS-CoV-2 may activate the in?ammasome

through mechanisms similar to those of SARS-CoV [91]. Overexpression of SARS-CoV-2

ORF3a in human cells can induce K

+

ef?ux, NLRP3 activation, and IL-1β release. Restricting

K

+

ef?ux with K

+

-rich media impairs the ability of ORF3a to trigger NLRP3 in?ammasome

Trends in Immunology, April 2022, Vol. 43, No. 4 289

Trends in Immunology

assembly, as evidenced from coimmunoprecipitation (co-IP) assays [92]. SARS-CoV-2 N

protein can interact directly with NLRP3, supported by results of reciprocal co-IP and confocal

microscopy, thus promoting in?ammasome activation and IL-1β release in cells. In Nlrp3

+/+

mice infected with adeno-associated virus (AAV)-Lung-N, serum IL-1β concentrations were in-

creased (detected via ELISA), whereas IL-1β was not induced by AAV-N in the sera of

Nlrp3

?/?

mice [93]. The higher cytokine concentrations stemming from NLRP3 activation

Trendsrends inin ImmunologyImmunology

Figure 2. Regulation oftheNLRP3in?ammasomebySARS-CoVinfection and NAD

+

. Severalproteinsencodedby

SARS-CoV promote NLRP3 in?ammasome activity and the release of proin?ammatory cytokines. ORF3a activates NF-κB

through TRAF3-dependent ubiquitination, which facilitates ASC speck formation and the assembly of NLRP3

in?ammasome [86]. ORF3a also has transmembrane domains and ion channel activity that drives a K

+

ef?ux [85].

E protein is located at the ER–Golgi compartment and promotes Ca

2+

ef?ux [87,88]. ORF8bdirectly interacts with

NLRP3. These mechanisms all activate the in?ammasome [89]. Host cell SIRT1, SIRT2, and SIRT3 all suppress the

NLRP3 in?ammsome [94–98]. SIRT1 deacetylates NF-κB, suppressing its activity, and reduces oxidative stress,

decreasing in?ammasome activation. SIRT2 deacetylates NLRP3. SIRT3 suppresses mitochondrial ROS production,

decreasing in?ammasome activation. SIRT6 reduces in?ammation via H3K9 deacetylation in the promoters of NF-κB

target genes [99]. PARPs promote antiviral SG formation through ADP-ribosylation of SG components [71–73].

Created with BioRender.com. Abbreviations: Ac, acetylation;ACE-2,angiotensin-convertingenzyme2;ASC,

apoptosis-associated speck-like protein containing a caspase recruitment domain Casp1, caspase-1; ER,

endoplasmic reticulum; NF-κB, nuclear factor κB; NLRP3, NOD-, LRR-, and pyrin domain-containing protein 3; ORF,

open reading frame; SARS-CoV, severe acute respiratory syndrome coronavirus 1; SIRT, sirtuin; TRAF3, TNF

receptor-associated factor 3; Ub, ubiquitination.

290 Trends in Immunology, April 2022, Vol. 43, No. 4

Trends in Immunology

suggest an increased likelihood of uncontrolled in?ammation in response to SARS-CoV-2

infection.

NAD

+

-consuming enzymes in anti-in?ammatory mechanisms

NAD

+

may contribute to the resolution of in?ammation, and to limiting or preventing the effects of

cytokine storms; it might do so by increasing the activity of sirtuins (Figure 2)[44]. SIRT1, SIRT2,

and SIRT3 all suppress the activity of NF-κB and the NLRP3 in?ammasome via multiple mecha-

nisms [94–98]. From a biochemical standpoint, SIRT1 physically interacts with and deacetylates

NF-κB and thereby suppressesitstranscriptionalactivity in humanepithelialcells[95]. SIRT1also

inhibits lipopolysaccharide-induced NLRP3 in?ammasome activation by reducing oxidative

stress, as demonstrated in human trophoblasts with an shRNA knockdown of SIRT1 [94].

SIRT2 directly deacetylates NLRP3 and inactivates the NLRP3 in?ammasome, as shown in

SIRT2 KO (Sirt2

-?

) mouse macrophages [97]. SIRT3 mediates in?ammasome activation by sup-

pressingmitochondrialreactiveoxygenspecies(ROS)production,whichcanactivatetheNLRP3

in?ammasome. SiRNA depletion of SIRT3 in human macrophages results in increased ROS

production and in?ammasome activation compared with controls [98].SIRT6alsopromotes

the resolution of in?ammation via histone H3 lysine 9 (H3K9) deacetylation in the promoters

of NF-κB target genes [99]. However, the effects of sirtuins on the NLRP3 in?ammasome have

yet to be extensively studied in the context of viral infection.

WepositthatNAD

+

boosters that suppress NF-κBandNLRP3in?ammasome activity might

also represent potential treatments to ameliorate the in?ammatory symptoms of COVID-19.

Indeed, treatment with NR, an NAD

+

precursor, for 24 hours has been reported to decrease

thereleaseofIL-1β and blunt in?ammasome activation in PBMCs from healthy and fasted

individuals [98]. NR also promotes SIRT1 expression, suppresses NLRP3 expression, and

reduces secretion of proin?ammatory factors TNFα and IL-6 in mouse hepatocytes [100].

Another NAD

+

precursor, nicotinic acid, also reduced in?ammation in a rodent model of type

2 diabetes (KK/HIJ mice) by modulating NLRP3 activity [26]. However, consideration should

be given to the fact that some studies show that extracellular nucleotides such as NAD

+

can

also promote in?ammation via the activation of P2X7 receptors in mouse macrophages and

T cells [101].

The promise of NAD

+

boosters in the clinic

Adding more fuel

Given that low NAD

+

concentrations might exacerbate COVID-19 severity, boosting the levels of

this metabolite in at-risk populations is one potential therapeutic strategy for mitigating severe

disease. The most straightforward way to raise NAD

+

concentrations is to provide additional pre-

cursors to boost NAD

+

synthesis [2]. Canonically, NAD

+

cannot be taken up directly by the cell,

but its precursors can [102,115]. The NAD

+

precursors nicotinic acid, NR, and NMN, all have pu-

tative transporters, high safety thresholds, and are orally bioavailable [2]. They do, however, vary

intheirpharmacokineticpro?lesandseemtoraiseNAD

+

concentrationstodifferentextentsindif-

ferent tissues [103], although this remains an area of active investigation.

Structurally, the closest molecule to NAD

+

is NMN, requiring only one enzymatic step to be con-

verted to NAD

+

. In a double blind, placebo-controlled trial, NMN was shown to increase insulin

sensitivityinprediabeticwomen [28] (NCT03151239;Table1). It iscurrently being studiedinhos-

pitalized COVID-19 patients with hypertension and metabolic syndrome for its effects on fatigue,

duration of hospitalization, and viral load (NCT05175768; Table 1). Ongoing clinical trials with

NMN are also investigating its effect on hypertension and postexercise muscle recovery

(NCT04903210 and NCT04664361; Table 1).

Trends in Immunology, April 2022, Vol. 43, No. 4 291

Outstanding questions

Why are NAD

+

tissue levels reduced

during aging? Many hypotheses exist

to date, but there is currently no

consensus, especially as multiple

mechanisms may be involved.

ArelowNAD

+

concentrations associated

with severe COVID-19? There is lit-

tle data to date showing a direct

association.

CanraisingNAD

+

tissueconcentrations

reduce COVID-19 symptoms, severity,

and mortality in the elderly and in the

young?

Can NAD

+

boosters reduce COVID-19

severity after the onset of symptoms,

or only prophylactically?

What might be the best approach for

raising NAD

+

concentrations in the

clinic: would this be NMN, NR, activa-

tors of NAD

+

biosynthesis, or inhibitors

of enzymes that consume NAD

+

?

What role does NAD

+

playinpromoting

SG-dependent antiviral mechanisms?

How does SARS-CoV-2 modulate

NAD

+

andNSP3toincreasevirulence?

Trends in Immunology

NR, which is two enzymatic steps removed from NAD

+

, is also being studied clinically. In one

placebo-controlled trial, oral NR reduced circulating concentrations of in?ammatory cytokines

IL-2, IL-5, and IL-6 [104] (NCT02950441; Table 1). In another trial, oral NR reduced blood pres-

sure and possibly aortic stiffness in healthy middle-aged and older adults [105] (NCT02921659;

Table 1). At the time this review was written, over 30 active studies testing NR had been regis-

tered on clinicaltrials.gov, including many for COVID-19 risk factors such as vascular disease,

hypertension, and heart failure (NCT04040959, NCT03821623, and NCT04528004; Table

1). At least four ongoing studies are directly testing NR in COVID-19 patients, some in patients

with particular risk factors such as advanced age or acute kidney injury, with readouts such as

rates of hospitalization, respiratory failure, and long-COVID symptoms (NCT04407390,

NCT04818216, NCT04573153, and NCT04809974; Table 1).

Other approaches to raising NAD

+

concentrations

Another approach to raising NAD

+

concentrations in humans is to inhibit the activity of NAD

+

-

consuming enzymes. For instance, as PARP1 is a major consumer of NAD

+

in cells [42], PARP1

inhibitors such as olaparib have been extensively studied for the treatment of breast and ovarian

cancer, given that PARP1 is synthetically lethal with BRCA mutations [106]. Regarding COVID-19,

several PARP1 inhibitors, CVL218 and stenoparib, were recently shown to limit SARS-CoV-2

replication and proin?ammatory cytokine production in human PBMCs and lung cells. One

hypothesized mechanism of action is via inhibition of PARP1-dependent NAD

+

depletion [107,116].

Several inhibitors of other NAD

+

-consuming enzymes are in preclinical development, including

against CD38 [108,109], SARM1 [110], and ACMSD (which consumes a precursor of NAD

+

in

the de novo pathway) [20]. In humans, treatment with luteolin, a naturally occurring CD38

inhibitor, reduced serum TNF and IL-6 concentrations [111].

The activation of NAD

+

biosynthetic enzymes is another strategy to raise NAD

+

concentrations; a

small molecule NAMPT activator, SBI-797812, is currently in preclinical development for cardio-

vascular and metabolic disease [112]. Finally, a virally‐encoded protein is a potential target that

interfereswithNAD

+

metabolism:themacrodomainofNSP3canantagonizetheantiviralactivities

of host PARPs,although the implications of this activity entail further investigation for SARS-CoV-

2[41,80–82]. A few inhibitors of NSP3 were recently identi?ed in a chemical screen and were

shown to exhibit antiviral properties against Chikungunya virus in human epithelial cells [113].

Concluding remarks

NAD

+

metabolism appears to be linked to infections of SARS-CoV-2 and other viruses via multiple

lines of evidence, epidemiological and mechanistic. Further research is warranted to better under-

stand its mechanistic role and utility as a target of intervention (see Outstanding questions). Clinical

translation of basic scienti?c discoveries is dif?cult, and there are many examples of antiviral and

anti-in?ammatory molecules that have shown early promise but have not materialized. However,

while human studies are still at an early stage, we are excited about the promise of interventions

thatmodulatetheconcentrationsofNAD

+

ortheactivityofNAD

+

-consumingenzymes.Suchinter-

ventions may soon have a place in our arsenal to ?ght current and future viral infections.

Declaration of interests

D.A.S. is a founder, equity owner, advisor to, director of, board member of, consultant to, investor in and/or inventor on

patents licensed to GlaxoSmithKline, Segterra, Animal Biosciences, AFAR, Cohbar, Galilei, Zymo Research, Immetas,

EdenRoc Sciences and af?liates (Arc-Bio, Dovetail Genomics, Claret Bioscience, MetroBiotech, Astrea, Liberty Biosecurity

and Delavie), Life Biosciences, and Levels Health. D.A.S. is an inventor on a patent application licensed to Elysium Health.

More at https://sinclair.hms.harvard.edu/david-sinclairs-af?liations.

292 Trends in Immunology, April 2022, Vol. 43, No. 4

Trends in Immunology

25. Trammell, S.A.J. et al. (2016) Nicotinamide riboside opposes

type 2 diabetes and neuropathy in mice. Sci. Rep. 6, 26933

49. Zingarelli, B. et al. (2003) Inhibitors of poly (ADP-ribose) poly-

merase modulate signal transduction pathways in colitis. Eur.

References

1. Stebbing, J. et al. (2020) COVID-19: combining antiviral and

anti-in?ammatory treatments. Lancet Infect. Dis. 20, 400–402

2. Rajman, L. et al. (2018) Therapeutic potential of NAD-boosting

molecules: the in vivo evidence. Cell Metab. 27, 529–547

3. Mueller, A.L. et al. (2020) Why does COVID-19 disproportion-

ately affect the elderly? Aging 12, 9959–9981

4. Zhou, F. et al. (2020) Clinical course and risk factors for mortal-

ity of adult inpatients with COVID-19 in Wuhan, China: a retro-

spective cohort study. Lancet 395, 1054–1062

5. Luk, J. et al. (2001) Observations on mortality during the 1918

in?uenza pandemic. Clin. Infect. Dis. 33, 1375–1378

6. Fang, E.F. et al. (2017) NAD+ in aging: molecular mechanisms

and translational implications. Trends Mol. Med. 23, 899–916

7. Gomes, A.P. et al. (2013) Declining NAD

+

induces a

pseudohypoxic state disrupting nuclear-mitochondrial com-

munication during aging. Cell 155, 1624–1638

8. Das, A. et al. (2018) Impairment of an endothelial NAD+-H2S

signaling network is a reversible cause of vascular aging. Cell

173, 74–89.e20

9. Camacho-Pereira, J. et al. (2016) CD38 dictates age-related

NAD decline and mitochondrial dysfunction through an

SIRT3-dependent mechanism. Cell Metab. 23, 1127–1139

10. Minhas, P.S. et al. (2019) Macrophage de novo NAD+ synthe-

sis speci?es immune function in aging and in?ammation. Nat.

Immunol. 20, 50–63

11. Clement, J. et al. (2019) The plasma NAD+ metabolome is dys-

regulated in “normal” aging. Rejuvenation Res. 22, 121–130

12. Massudi, H. et al. (2012) Age-associated changes in oxidative

stress and NAD+ metabolism in human tissue. PLoS One 7,

e42357

13. Zhou, C.-C. et al. (2016) Hepatic NAD(+) de?ciency as a thera-

peutic target for non-alcoholic fatty liver disease in ageing. Br.

J. Pharmacol. 173, 2352–2368

14. Essuman, K. et al. (2017) The SARM1 Toll/interleukin-1 recep-

tor domain possesses intrinsic NAD+ cleavage activity that

promotes pathological axonal degeneration. Neuron 93,

1334–1343.e5

15. Bai, P. et al. (2011) PARP-1 inhibition increases mitochondrial

metabolism through SIRT1 activation. Cell Metab. 13, 461–468

16. Li, J. et al. (2017) A conserved NAD+ binding pocket that reg-

ulates protein-protein interactions during aging. Science 355,

1312–1317

17. Covarrubias, A.J. et al. (2020) Senescent cells promote tissue

NAD+ decline during ageing via the activation of CD38+

macrophages. Nat. Metab. 2, 1265–1283

18. Braidy, N. et al. (2011) Effects of kynurenine pathway inhibition

on NAD metabolism and cell viability in human primary astro-

cytes and neurons. Int. J. Tryptophan Res. 4, 29–37

19. Frederick, D.W. et al. (2016) Loss of NAD homeostasis leads to

progressive and reversible degeneration of skeletal muscle.

Cell Metab. 24, 269–282

20. Katsyuba, E. et al. (2018) De novo NAD

+

synthesis enhances

mitochondrial function and improves health. Nature 563,

354–359

21. Zhu, L. et al. (2020) Association of blood glucose control and

outcomes in patients with COVID-19 and pre-existing type 2

diabetes. Cell Metab. 31, 1068–1077.e3

22. Cantó, C. et al. (2012) The NAD(+) precursor nicotinamide

riboside enhances oxidative metabolism and protects against

high-fat diet-induced obesity. Cell Metab. 15, 838–847

23. Yoshino, J. et al. (2011) Nicotinamide mononucleotide, a key

NAD(+) intermediate, treats the pathophysiology of diet- and

age-induced diabetes in mice. Cell Metab. 14, 528–536

24. Mills, K.F. et al. (2016) Long-term administration of nicotin-

amide mononucleotide mitigates age-associated physiological

decline in mice. Cell Metab. 24, 795–806

discordant monozygotic twins. J. Clin. Endocrinol. Metab.

101, 275–283

28. Yoshino,M.etal.(2021)Nicotinamidemononucleotideincreases

muscle insulin sensitivity in prediabetic women. Science 372,

1224–1229

29. Hallakou-Bozec, S. et al. (2021) Mechanism of action of

Imeglimin: a novel therapeutic agent for type 2 diabetes.

Diabetes Obes. Metab. 23, 664–673

30. Xu, J. et al. (2021) A meta-analysis on the risk factors adjusted

association between cardiovascular disease and COVID-19

severity. BMC Public Health 21, 1533

31. de Picciotto, N.E. et al. (2016) Nicotinamide mononucleotide

supplementation reverses vascular dysfunction and oxidative

stress with aging in mice. Aging Cell 15, 522–530

32. Tong, D. et al. (2021) NAD+ repletion reverses heart failure with

preserved ejection fraction. Circ. Res. 128, 1629–1641

33. Kane, A.E. and Sinclair, D.A. (2018) Sirtuins and NAD+ in the

development and treatment of metabolic and cardiovascular

diseases. Circ. Res. 123, 868–885

34. Kaplon, R.E. et al. (2014) Vascular endothelial function and ox-

idative stress are related to dietary niacin intake among healthy

middle-aged and older adults. J. Appl. Physiol. 116, 156–163

35. Murray, M.F. et al. (1995) HIV infection decreases intracellular

nicotinamide adenine dinucleotide [NAD]. Biochem. Biophys.

Res. Commun. 212, 126–131

36. Grady, S.L. et al. (2012) Herpes simplex virus 1 infection

activates poly(ADP-ribose) polymerase and triggers the degra-

dation of poly(ADP-ribose) glycohydrolase. J. Virol. 86,

8259–8268

37. Tran, T. et al. (2022) Reduced levels of NAD in skeletal muscle

and increased physiologic frailty are associated with viral coin-

fection in asymptomatic middle-aged adults. J. Acquir.

Immune De?c. Syndr. 89, S15–S22

38. Maurya, S.P. et al. (2019) Effect of Withania somnifer on CD38

expression on CD8+ T lymphocytes among patients of HIV

infection. Clin. Immunol. 203, 122–124

39. Xiao, N. et al. (2021) Integrated cytokine and metabolite analysis

reveals immunometabolic reprogramming in COVID-19 patients

with therapeutic implications. Nat. Commun. 12, 1618

40. Heer, C.D. et al. (2020) Coronavirus infection and PARP expres-

sion dysregulate the NAD metabolome: an actionable component

of innate immunity. J. Biol. Chem. 295, 17986–17996

41. Grunewald, M.E. et al. (2019) The coronavirus macrodomain is

required to prevent PARP-mediated inhibition of virus replica-

tion and enhancement of IFN expression. PLoS Pathog. 15,

e1007756

42. Brady, P.N. et al. (2019) Poly(ADP-Ribose) polymerases in

host-pathogen interactions, in?ammation, and immunity.

Microbiol. Mol. Biol. Rev. 83, e00038-18

43. Shang, J. et al. (2021) NAD+-consuming enzymes in immune

defense against viral infection. Biochem. J. 478, 4071–4092

44. Verdin, E. (2015) NAD

+

in aging, metabolism, and neurodegen-

eration. Science 350, 1208–1213

45. Boulares, A.H. et al. (2003) Gene knockout or pharmacological

inhibition of poly(ADP-ribose) polymerase-1 prevents lung

in?ammation in a murine model of asthma. Am. J. Respir. Cell

Mol. Biol. 28, 322–329

46. Suzuki, Y. et al. (2004) Inhibition of poly(ADP-ribose) polymerase

prevents allergen-induced asthma-like reaction in sensitized

Guinea pigs. J. Pharmacol. Exp. Ther. 311, 1241–1248

47. Jijon, H.B. et al. (2000) Inhibition of poly(ADP-ribose) polymer-

ase attenuates in?ammation in a model of chronic colitis. Am.

J. Physiol. Gastrointest. Liver Physiol. 279, G641–G651

48. Larmonier, C.B. et al. (2016) Transcriptional reprogramming and

resistancetocolonicmucosalinjuryinpoly(ADP-ribose)polymer-

ase 1 (PARP1)-de?cient mice. J. Biol. Chem. 291, 8918–8930

26. Lee, H.J. et al. (2015) Nicotinamide riboside ameliorates hepatic

meta?ammation by modulating NLRP3 in?ammasome in a

rodent model of type 2 diabetes. J. Med. Food 18, 1207–1213

27. Jukarainen, S. et al. (2016) Obesity is associated with low

NAD(+)/SIRT pathway expression in adipose tissue of BMI-

J. Pharmacol. 469, 183–194

50. Altmeyer, M. et al. (2010) Absence of poly(ADP-ribose) poly-

merase 1 delays the onset of Salmonella enterica serovar

Typhimurium-induced gut in?ammation. Infect. Immun. 78,

3420–3431

Trends in Immunology, April 2022, Vol. 43, No. 4 293

Trends in Immunology

51. Czapski, G.A. et al. (2006) Poly(ADP-ribose) polymerase-1

inhibition protects the brain against systemic in?ammation.

Neurochem. Int. 49, 751–755

52. Atasheva, S. et al. (2012) New PARP gene with an anti-

alphavirus function. J. Virol. 86, 8147–8160

53. Goodier, J.L. et al. (2015) The broad-spectrum antiviral protein

ZAP restricts human retrotransposition. PLoS Genet. 11,

e1005252

54. Zhu, Y. et al. (2011) Zinc-?nger antiviral protein inhibits

HIV-1 infection by selectively targeting multiply spliced

viral mRNAs for degradation. Proc.Natl.Acad.Sci.U.S.A.

108, 15834–15839

55. Müller, S. et al. (2007) Inhibition of ?lovirus replication by the

zinc ?nger antiviral protein. J. Virol. 81, 2391–2400

56. Mao, R. et al. (2013) Inhibition of hepatitis B virus replication by

the host zinc ?nger antiviral protein. PLoS Pathog. 9, e1003494

57. Grunewald, M.E. et al. (2018) The coronavirus nucleocapsid

protein is ADP-ribosylated. Virology 517, 62–68

58. Iwata, H. et al. (2016) PARP9 and PARP14 cross-regulate

macrophage activation via STAT1 ADP-ribosylation. Nat.

Commun. 7, 12849

59. Atasheva,S.etal.(2014) Interferon-stimulated poly(ADP-Ribose)

polymerases are potent inhibitors of cellular translation and virus

replication. J. Virol. 88, 2116–2130

60. Das, D. et al. (2017) The deacetylase SIRT1 regulates the rep-

lication properties of human papillomavirus 16 E1 and E2.

J. Virol. 91, 00102–00117

61. Campagna, M. et al. (2011) SIRT1 stabilizes PML promoting its

sumoylation. Cell Death Differ. 18, 72–79

62. Li, Q. et al. (2014) Activation of Kaposi’s sarcoma-associated

herpesvirus (KSHV) by inhibitors of class III histone

deacetylases: identi?cation of sirtuin 1 as a regulator of the

KSHV life cycle. J. Virol. 88, 6355–6367

63. Koyuncu, E. et al. (2014) Sirtuins are evolutionarily conserved

viral restriction factors. MBio 5, e02249-14

64. Jiang, H. et al. (2013) SIRT6 regulates TNF-α secretion through

hydrolysis of long-chain fatty acyl lysine. Nature 496, 110–113

65. Dantoft, W. et al. (2019) Metabolic regulators nampt and sirt6

seriallyparticipateinthemacrophageinterferonantiviralcascade.

Front. Microbiol. 10, 355

66. Horenstein, A.L. et al. (2021) CD38 in the age of COVID-19: a

medical perspective. Physiol. Rev. 101, 1457–1486

67. Hernández-Campo, P.M. et al. (2006) Normal patterns of

expression of glycosylphosphatidylinositol-anchored proteins

ondifferentsubsetsofperipheralbloodcells:aframeofreference

for the diagnosis of paroxysmal nocturnal hemoglobinuria.

Cytometry B Clin. Cytom. 70, 71–81

68. Figley, M.D. and DiAntonio, A. (2020) The SARM1 axon degen-

eration pathway: control of the NAD+ metabolome regulates

axon survival in health and disease. Curr. Opin. Neurobiol.

63, 59–66

69. McCormick, C. and Khaperskyy, D.A. (2017) Translation inhibi-

tion and stress granules in the antiviral immune response. Nat.

Rev. Immunol. 17, 647–660

70. Raaben, M. et al. (2007) Mouse hepatitis coronavirus replica-

tion induces host translational shutoff and mRNA decay, with

concomitant formation of stress granules and processing

bodies. Cell. Microbiol. 9, 2218–2229

71. Leung, A.K.L. et al. (2011) Poly(ADP-ribose) regulates stress

responses and microRNA activity in the cytoplasm. Mol. Cell

42, 489–499

72. Patel, A. et al. (2015) A liquid-to-solid phase transition of the

ALS protein FUS accelerated by disease mutation. Cell 162,

1066–1077

73. Altmeyer, M. et al. (2015) Liquid demixing of intrinsically disor-

dered proteins is seeded by poly(ADP-ribose). Nat. Commun.

6, 8088

74. Saikatendu, K.S. et al. (2005) Structural basis of severe acute

respiratory syndrome coronavirus ADP-ribose-1″-phosphate

dephosphorylation by a conserved domain of nsP3. Structure

76. Abraham, R. et al. (2018) ADP-ribosyl-binding and hydrolase

activities of the alphavirus nsP3 macrodomain are critical for

initiation of virus replication. Proc.Natl.Acad.Sci.U.S.A.

115, E10457–E10466

77. Alhammad, Y.M.O. and Fehr, A.R. (2020) The viral macro-

domain counters host antiviral ADP-ribosylation. Viruses

12, 384

78. Minakshi, R. and Padhan, K. (2014) The YXXΦ motif within the

severe acute respiratorysyndrome coronavirus (SARS-CoV) 3a

protein is crucial for its intracellular transport. Virol. J. 11, 75

79. Frick, D.N. et al. (2020) Molecular basis for ADP-ribose binding

to the Mac1 domain of SARS-CoV-2 nsp3. Biochemistry 59,

2608–2615

80. Alhammad, Y.M.O. et al. (2021) The SARS-CoV-2 conserved

macrodomain is a mono-ADP-ribosylhydrolase. J. Virol. 95,

01969-20

81. Russo, L.C. et al. (2021) The SARS-CoV-2 Nsp3 macrodomain

reverses PARP9/DTX3L-dependent ADP-ribosylation induced

by interferon signaling. J. Biol. Chem. 297, 101041

82. Fehr, A.R. et al. (2016) The conserved coronavirus

macrodomain promotes virulence and suppresses the innate

immune response during severe acute respiratory syndrome

coronavirus infection. MBio 7, e01721-16

83. Herold, T. et al. (2020) Elevated levels of IL-6 and CRP predict

the need for mechanical ventilation in COVID-19. J. Allergy Clin.

Immunol. 146, 128–136.e4

84. Huang, C. et al. (2020) Clinical features of patients infected with

2019 novel coronavirus in Wuhan, China. Lancet 395,

497–506

85. Chen, I.-Y. et al. (2019) Severe acute respiratory syndrome co-

ronavirus viroporin 3a activates the NLRP3 in?ammasome.

Front. Microbiol. 10, 50

86. Siu, K.-L. et al. (2019) Severe acute respiratory syndrome co-

ronavirus ORF3a protein activates the NLRP3 in?ammasome

by promoting TRAF3-dependent ubiquitination of ASC.

FASEB J. 33, 8865–8877

87. Casta?o-Rodriguez, C. et al. (2018) Role of severe acute

respiratory syndrome coronavirus viroporins E, 3a, and 8a in

replication and pathogenesis. MBio 9, e02325-17

88. Nieto-Torres, J.L. et al. (2015) Severe acute respiratory

syndrome coronavirus E protein transports calcium ions and

activates the NLRP3 in?ammasome. Virology 485, 330–339

89. Shi, C.-S. et al. (2019) SARS-coronavirus open reading frame-

8b triggers intracellular stress pathways and activates NLRP3

in?ammasomes. Cell Death Discov. 5, 101

90. Nieto-Torres, J.L. et al. (2014) Severe acute respiratory

syndrome coronavirus envelope protein ion channel activity

promotes virus ?tness and pathogenesis. PLoS Pathog. 10,

e1004077

91. Rodrigues, T.S. et al. (2021) In?ammasomes are activated in

response to SARS-CoV-2 infection and are associated with

COVID-19 severity in patients. J. Exp. Med. 218, e20201707

92. Xu, H. et al. (2022) SARS-CoV-2 viroporin encoded by ORF3a

triggers the NLRP3 in?ammatory pathway. Virology 568, 13–22

93. Pan, P. et al. (2021) SARS-CoV-2 N protein promotes NLRP3

in?ammasome activation to induce hyperin?ammation. Nat.

Commun. 12, 4664

94. Park, S. et al. (2020) SIRT1 Alleviates LPS-induced IL-1β pro-

duction by suppressing NLRP3 in?ammasome activation and

ROS production in trophoblasts. Cells 9, 3

95. Yeung, F. et al. (2004) Modulation of NF-kappaB-dependent

transcription and cell survival by the SIRT1 deacetylase.

EMBO J. 23, 2369–2380

96. Li, Y. et al. (2017) Negative regulation of NLRP3 in?ammasome

by SIRT1 in vascular endothelial cells. Immunobiology 222,

552–561

97. He, M. et al. (2020) An acetylation switch of the NLRP3 in?am-

masome regulates aging-associated chronic in?ammation and

insulin resistance. Cell Metab. 31, 580–591.e5

98. Traba, J. et al. (2015) Fasting and refeeding differentially regu-

13, 1665–1675

75. Jayabalan, A.K. et al. (2021) Stress granule formation, disas-

sembly, and composition are regulated by alphavirus ADP-

ribosylhydrolase activity. Proc. Natl. Acad. Sci. U. S. A. 118,

e2021719118

294 Trends in Immunology, April 2022, Vol. 43, No. 4

late NLRP3 in?ammasome activation in human subjects.

J. Clin. Invest. 125, 4592–4600

99. Kawahara, T.L.A. et al. (2009) SIRT6 links histone H3 lysine 9

deacetylation to NF-kappaB-dependent gene expression and

organismal life span. Cell 136, 62–74

100. Lee,H.J. and Yang, S.J.(2019) Nicotinamide riboside regulates

in?ammation and mitochondrial markers in AML12 hepatocytes.

Nutr. Res. Pract. 13, 3–10

101. Hong, S. et al. (2009) Differential regulation of P2X7 receptor

activation by extracellular nicotinamide adenine dinucleotide

and ecto-ADP-ribosyltransferases in murine macrophages

and T cells. J. Immunol. 183, 578–592

102. Grozio, A. et al. (2019) Slc12a8 is a nicotinamide mononucleo-

tide transporter. Nat. Metab. 1, 47–57

103. Liu, L. et al. (2018) Quantitative analysis of NAD synthesis-

breakdown ?uxes. Cell Metab. 27, 1067–1080.e5

104. Elhassan,Y.S.et al.(2019)Nicotinamideribosideaugmentstheaged

human skeletal muscle NAD+ metabolome and induces transcripto-

mic and anti-in?ammatory signatures. Cell Rep. 28, 1717–1728.e6

105. Martens, C.R. et al. (2018) Chronic nicotinamide riboside sup-

plementation is well-tolerated and elevates NAD+ in healthy

middle-aged and older adults. Nat. Commun. 9, 1286

106. Farmer, H. et al. (2005) Targeting the DNA repair defect in BRCA

mutant cells as a therapeutic strategy. Nature 434, 917–921

107. Ge, Y. et al. (2021) An integrative drug repositioning framework

discovered a potential therapeutic agent targeting COVID-19.

Signal Transduct. Target. Ther. 6, 165

108. Haffner, C.D. et al. (2015) Discovery, synthesis, and biological

evaluation ofthiazoloquin(az)olin(on)es aspotent CD38inhibitors.

J. Med. Chem. 58, 3548–3571

109. Tarragó, M.G. et al. (2018) A potent and speci?c CD38 inhibitor

ameliorates age-related metabolic dysfunction by reversing tis-

sue NAD+ decline. Cell Metab. 27, 1081–1095.e10

110. Loring, H.S. et al. (2020) Identi?cation of the ?rst noncompeti-

tive SARM1 inhibitors. Bioorg. Med. Chem. 28, 115644

111. Tsilioni, I. et al. (2015) Children with autism spectrum disorders,

who improved with a luteolin-containing dietary formulation,

show reduced serum levels of TNF and IL-6. Transl. Psychiatry

5, e647

112. Gardell, S.J. et al. (2019) Boosting NAD+ with a small molecule

that activates NAMPT. Nat. Commun. 10, 3241

113. Shimizu, J.F. et al. (2020) Is the ADP ribose site of the

Chikungunya virus NSP3 Macro domain a target for antiviral

approaches? Acta Trop. 207, 105490

114. Kropotov, A. et al. (2021) Equilibrative nucleoside transporters

mediate the import of nicotinamide riboside and nicotinic acid

riboside into human cells. Int. J. Mol. Sci. 22, 1391

115. Stone, N.E. et al. (2021) Stenoparib, an inhibitor of cellular poly

(ADP-ribose) polymerase, blocks replication of the SARS-CoV-

2 and HCoV-NL63 human coronaviruses in vitro. mBio 12,

e3495-20

116. Janssens, G.E. et al. (2022) Healthy aging and muscle function

are positively associated with NAD

+

abundance in humans.

Nat. Aging Published online February 17, 2022. https://doi.

org/10.1038/s43587-022-00174-3

Trends in Immunology

Trends in Immunology, April 2022, Vol. 43, No. 4 295

献花(0)
+1
(本文系jlc108首藏)