Modulation of innate immunity of patients with Alzheimer’s ... · THE JOURNAL † REVIEW †...

12
THE JOURNAL REVIEW www.fasebj.org Modulation of innate immunity of patients with Alzheimers disease by omega-3 fatty acids Milan Fiala,* ,1 Gijs Kooij, ,Karen Wagner, § Bruce Hammock, § and Matteo Pellegrini* *Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, California, USA; Department of Molecular Cell Biology and Immunology, Vrije Universiteit (VU) Medical Center, Multiple Sclerosis Center Amsterdam, Amsterdam, The Netherlands; Perioperative and Pain Medicine, Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Harvard Institutes of Medicine, Brigham and Womens Hospital and Harvard Medical School, Boston, Massachusetts, USA; and § Department of Entomology, University of California, Davis, Davis, California, USA ABSTRACT: The innate immune system of patients with Alzheimers disease and mild cognitive impairment (MCI) is deregulated with highly increased or decreased transcription of inflammatory genes and consistently depressed phagocytosis of amyloid-b 1-42 (Ab) by monocytes and macrophages. Current immune therapies target single mechanisms in the adaptive immune system but not innate immunity. Here, we summarize recent advances in therapy by v-3, v-6, and epoxy fatty acids; specialized proresolving mediators; and vitamin D 3 that have proven immune effects and emerging cognitive effects in patients with MCI. The hypothesis of this approach is that macrophages of normal participants, but not those of patients with Alzheimers disease and MCI, possess effective phagocytosis for Ab and protect homeostasis of the brain and, furthermore, that defective MCI macrophages recover phagocytic function via v-3. Recent studies of fish-derived v-3 supplementation in patients with MCI have shown polarization of Apo«3/«3 patientsmacrophages to an intermediate M1-M2 phenotype that is optimal for Ab phagocytosis and the stabilization of cognitive decline. Therefore, accumulating preclinical and preliminary clinical evidence indicates that v-3 supplementation should be tested in a randomized controlled clinical trial and that the analysis should involve the apolipoprotein E genotype and intervening conditions during trial. Fiala, M., Kooij, G., Wagner, K., Hammock, B., Pellegrini, M. Modulation of innate immunity of patients with Alzheimers disease by omega-3 fatty acids. FASEB J. 31, 000000 (2017). www.fasebj.org Alzheimers disease (AD) is a neurodegenerative disease that is preceded by prodromal stages of mild cognitive impairment (MCI) (1) and subjective cognitive impair- ment (2) that frequently are not recognized but are im- portant for the start of preventive therapy. To date, there is no proven effective therapy to counteract the progression of MCI to AD, which has a 42.5% incidence at 5 yr (3); however, it is accepted that any therapy should address inflammatory damage and clearance of amyloid-b 1-42 (Ab) (4). Potential therapeutics include v-3 fatty acidderived proresolving mediators, which induce anti- inflammatory and proresolving responses and enhance macrophage phagocytosis (5), including the phagocytosis of Ab (6). In addition, the v-6 fatty acid arachidonic acidderived mediator, epoxyeicosatrienoic acid (EET), displays potent anti-inflammatory and antinociceptive properties (7). Recent discoveries that have bearing on the transcriptional deregulation of innate immunity against Ab (6) and alterations in resolution pathways in the CNS of patients with AD (8) suggest that v-3 lipid mediators should be tested in a controlled trial. MACROPHAGES AND MICROGLIA IN THE AD BRAIN: HARMFUL INFLAMMATION AND BENEFICIAL CLEARANCE OF Ab Roles in AD of microglia- vs. monocyte-derived macro- phages have been discussed since the 90s. Microglia that originate from bone marrow were shown to clear Ab (9) but also to have toxic effects on neurons (10). Inflammation in the AD brain was attributed to Ab deposits that had been invaded by microglia and surrounded by reactive astrocytes (11). Monocytes were attracted into neuropil across the human blood-brain barrier (BBB) model by CCL2, -3, and -4 chemokines (12) and, in mouse models, by ABBREVIATIONS: 1,25D 3 ,1a,25-dihydroxy vitamin D 3 ;Ab, amyloid-b 1-42 ; AD, Alzheimers disease; APOE, apolipoprotein E; AT, aspirin-triggered; BBB, blood-brain barrier; DHA, docosahexaenoic acid; EET, epoxy- eicosatrienoic acid; EPA, eicosapentaenoic acid; EpFA, epoxy fatty acid; GlcNAc, b-1,4-mannosyl-glycoprotein4-b-N-acetylglucosaminyltransfer- ase; IRF, IFN regulatory factor; LXA 4 , lipoxin A 4 ; MaR1, maresin 1; MCI, mild cognitive impairment; MMSE, minimental state examination; PPAR, peroxisome proliferator-activated receptor; PUFA, polyunsaturated fatty acid; RvD, resolvin D; RXR, retinoid X receptor; sHE, soluble epoxide hydrolase; SPM, specialized proresolving mediator; STAT, signal trans- ducer and activator of transcription 1 Correspondence: UCLA School of Medicine, 100 UCLA Medical Plaza, Suite 220, Los Angeles, CA 90095, USA. E-mail: [email protected] doi: 10.1096/fj.201700065R 0892-6638/17/0031-0001 © FASEB 1 The FASEB Journal article fj.201700065R. Published online April 18, 2017. Vol., No. , pp:, April, 2017 The FASEB Journal . 169.237.169.81 to IP www.fasebj.org Downloaded from

Transcript of Modulation of innate immunity of patients with Alzheimer’s ... · THE JOURNAL † REVIEW †...

Page 1: Modulation of innate immunity of patients with Alzheimer’s ... · THE JOURNAL † REVIEW † Modulation of innate immunity of patients with Alzheimer’s disease by omega-3 fatty

THE

JOURNAL • REVIEW • www.fasebj.org

Modulation of innate immunity of patients withAlzheimer’s disease by omega-3 fatty acidsMilan Fiala,*,1 Gijs Kooij,†,‡ Karen Wagner,§ Bruce Hammock,§ and Matteo Pellegrini**Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, California, USA; †Departmentof Molecular Cell Biology and Immunology, Vrije Universiteit (VU) Medical Center, Multiple Sclerosis Center Amsterdam, Amsterdam, TheNetherlands; ‡Perioperative and Pain Medicine, Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology,Harvard Institutes of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA; and §Departmentof Entomology, University of California, Davis, Davis, California, USA

ABSTRACT:The innate immune systemof patientswithAlzheimer’s disease andmild cognitive impairment (MCI) isderegulated with highly increased or decreased transcription of inflammatory genes and consistently depressedphagocytosis of amyloid-b1-42 (Ab) by monocytes and macrophages. Current immune therapies target singlemechanisms in the adaptive immune system but not innate immunity. Here, we summarize recent advances intherapy by v-3, v-6, and epoxy fatty acids; specialized proresolving mediators; and vitamin D3 that have provenimmune effects and emerging cognitive effects in patients with MCI. The hypothesis of this approach is thatmacrophages of normal participants, but not those of patientswith Alzheimer’s disease andMCI, possess effectivephagocytosis forAb andprotecthomeostasisof thebrainand, furthermore, thatdefectiveMCImacrophages recoverphagocytic function viav-3. Recent studies of fish-derivedv-3 supplementation in patientswithMCI have shownpolarization of Apo«3/«3 patients’ macrophages to an intermediate M1-M2 phenotype that is optimal for Abphagocytosis and the stabilizationof cognitivedecline.Therefore, accumulatingpreclinical andpreliminary clinicalevidence indicates thatv-3 supplementation should be tested in a randomized controlled clinical trial and that theanalysis should involve the apolipoprotein E genotype and intervening conditions during trial.—Fiala, M., Kooij,G.,Wagner,K.,Hammock,B.,Pellegrini,M.Modulationof innate immunityofpatientswithAlzheimer’sdiseasebyomega-3 fatty acids. FASEB J. 31, 000–000 (2017). www.fasebj.org

Alzheimer’s disease (AD) is a neurodegenerative diseasethat is preceded by prodromal stages of mild cognitiveimpairment (MCI) (1) and subjective cognitive impair-ment (2) that frequently are not recognized but are im-portant for the start of preventive therapy. To date, there isno proven effective therapy to counteract the progressionof MCI to AD, which has a 42.5% incidence at 5 yr (3);however, it is accepted that any therapy should addressinflammatory damage and clearance of amyloid-b1-42(Ab) (4). Potential therapeutics include v-3 fatty acid–derived proresolving mediators, which induce anti-

inflammatory and proresolving responses and enhancemacrophage phagocytosis (5), including the phagocytosisof Ab (6). In addition, the v-6 fatty acid arachidonicacid–derived mediator, epoxyeicosatrienoic acid (EET),displays potent anti-inflammatory and antinociceptiveproperties (7). Recent discoveries that have bearing on thetranscriptional deregulation of innate immunity againstAb (6) and alterations in resolution pathways in the CNSof patients with AD (8) suggest that v-3 lipid mediatorsshould be tested in a controlled trial.

MACROPHAGES AND MICROGLIA IN THE ADBRAIN: HARMFUL INFLAMMATION ANDBENEFICIAL CLEARANCE OF Ab

Roles in AD of microglia- vs. monocyte-derived macro-phages have been discussed since the 90s. Microglia thatoriginate from bone marrow were shown to clear Ab (9)but also tohave toxic effects onneurons (10). Inflammationin the AD brain was attributed to Ab deposits that hadbeen invaded by microglia and surrounded by reactiveastrocytes (11). Monocytes were attracted into neuropilacross the human blood-brain barrier (BBB) model byCCL2, -3, and -4 chemokines (12) and, inmousemodels, by

ABBREVIATIONS: 1,25D3, 1a,25-dihydroxy vitamin D3; Ab, amyloid-b1-42;AD, Alzheimer’s disease; APOE, apolipoprotein E; AT, aspirin-triggered;BBB, blood-brain barrier; DHA, docosahexaenoic acid; EET, epoxy-eicosatrienoic acid; EPA, eicosapentaenoic acid; EpFA, epoxy fatty acid;GlcNAc, b-1,4-mannosyl-glycoprotein4-b-N-acetylglucosaminyltransfer-ase; IRF, IFN regulatory factor; LXA4, lipoxin A4; MaR1, maresin 1; MCI,mild cognitive impairment; MMSE, minimental state examination; PPAR,peroxisome proliferator-activated receptor; PUFA, polyunsaturated fattyacid; RvD, resolvin D; RXR, retinoid X receptor; sHE, soluble epoxidehydrolase; SPM, specialized proresolving mediator; STAT, signal trans-ducer and activator of transcription1 Correspondence: UCLA School of Medicine, 100 UCLA Medical Plaza,Suite 220, Los Angeles, CA 90095, USA. E-mail: [email protected]

doi: 10.1096/fj.201700065R

0892-6638/17/0031-0001 © FASEB 1

The FASEB Journal article fj.201700065R. Published online April 18, 2017.

 Vol.,  No. , pp:, April, 2017The FASEB Journal. 169.237.169.81 to IP www.fasebj.orgDownloaded from

Page 2: Modulation of innate immunity of patients with Alzheimer’s ... · THE JOURNAL † REVIEW † Modulation of innate immunity of patients with Alzheimer’s disease by omega-3 fatty

CCR2 (13). Subsequently, transplantation of bonemarrowand bone marrow–derived microglia were tested as ther-apy for neurodegenerative diseases in animalmodels andin humans, including in patients with multiple sclerosis(14); however, further discoveries have complicated thisapproach, as the origin of adultmicrogliawas shown to beprenatal from primitive myeloid progenitors in the yolksac (15).Meanwhile, in studies relevant toAD, invasion ofmonocyte-derived macrophages was noticed in the hu-man brain with ischemic plaques (16) and the AD brain(17). The investigation of brain macrophages by RNA se-quencing in a mouse model has shown that CNS macro-phages arise from yolk sac precursors during embryonicdevelopment and are not significantly related to circulat-ing myeloid cells but are activated during inflammation(18, 19). However, the inflammatory model in the animalstudy (19) has uncertain relation to monocyte and/ormacrophage infiltration of the AD brain. An extensivebody of literature shows monocyte migration across theBBB in HIV-1 encephalitis (20) that histopathologicallyresembles the AD brain (17). In an in vitro model, mono-cytes are attracted by Ab-induced chemokines to migrateacross the BBB (12).

Nonetheless, in mouse models, the role of monocyte-derivedmacrophages has remained controversial becausemonocyte invasion into theCNSwas found tobe related toradiation- or chemotherapy-inducedBBBdamage (21, 22).With the exception of patrolling circulating monocytesthat are chargedwith clearing vascularAb (23), the role ofAb clearance has been ascribed to microglia (24). How-ever, there has also been strong evidence for peripheralmonocytes playing a role in Ab clearance: 1) Ab degra-dation by mouse microglia was inefficient (25); 2) in-terference with monocyte migration by CCR2 deficiencyresulted in increased levels of cerebral Ab deposition andcognitive impairment (13); 3) CCR2 deficiency of peri-vascularmyeloid cells impairedAb clearance (26); 4) PD-1immune checkpoint blockade and regulatory T cell sup-pression increased recruitment of monocyte-derivedmacrophages in a mouse brain and cleared Ab (27, 28);and 5) in experimental autoimmune encephalitis, the ef-fective antigen presenting cells are CD45highCD11b/c+

transitional macrophages, not CD45lowCD11b/c+ micro-glia (29). Microglia are believed to clear Ab-coated Ab viathe Ab Fc domain (30, 31), but the effectiveness of Fc im-mune clearance is contradicted by poor results of Ab Abclearance (32).Conversely, therehasbeenevidence againstAb clearance by monocytes and/or macrophages in themouse model, as rapid repopulation of the brain withperipheral myeloid cells, even in the presence of anti-AbAb, failed to clear Ab plaques (33, 34).

The genetic mouse model that overexpresses theamyloid-precursor protein with pathogenic mutationsdoes not express the pathognomonic immunedefect ofAbphagocytosis bymacrophages of patients withAD (35). Inaddition, the mouse response to 1a,25-dihydroxy vitaminD3 (1,25D3) is different compared with the human re-sponse (36), and, in general, inflammatory responsesdifferbetween mice and humans (37). Finally, immune activa-tion of the hNOS2 gene is crucial in an animal modelto mimic the pathologic phenotype of AD (38). The

examination of brain tissues and immune cells fromhuman patients with AD demonstrates that the immu-nopathology and clearance of Ab depend upon mono-cytes andmacrophages (17). In theADbrain tissue assay,normal monocytes, but not AD monocytes, clear Abfromneurons (35).Monocytes that had phagocytizedAbappear fat and do not transmigrate across the BBB,whereas lean monocytes do. AD macrophages are de-fective in Ab phagocytosis and degradation and areprone to apoptosis from Ab (39); thus, in the healthybrain, monocytes may immigrate into the neuropil, clearAb by phagocytosis, and emigrate, whereas in the ADbrain, AD monocytes may not. This hypothesis is sup-ported by a study of the AD brain that shows apoptoticmacrophages that contain fibrillary Ab on the abluminalside of microvessels that circumferentially surround thecongophilic vessel wall and also colocalize with neurons(39) and plaques (17) (Fig. 1). In some patients, increasedexpression ofCD33 is associatedwith the rs3865444C riskallele for AD and decreased Ab clearance (40, 41). Theseconsiderations suggest that peripheral blood monocytesare diagnostic and therapeutic tools.

Early epidemiologic studies have pointed to the role ofinflammation because the prevalence of AD was reducedin patients who were treated with nonsteroidal anti-inflammatory drugs (42) and the brain showed the pres-ence of membrane attack complex and T cells (43). Theroles of the initiating factor of the classic complementcascade, C1q and C3 component, have been revisited inmouse models in which they were found to be associatedwith early synapse loss (44). In a recent study, macro-phages of most patients with ADdisplayed inflammatorymarkers, including CD14, TLR 2, TLR4, IL6, and CCR2, aswell asMHC-II/Ab complexes (45). Exposureofmicrogliatodocosahexaenoic acid (DHA)andeicosapentaenoic acid(EPA) decreased inflammatoryM1markers and increasedAb phagocytosis and anti-inflammatoryM2markers (46).Translation of these results to patients with MCI is notstraightforward because the inflammatory activation ofperipheral blood mononuclear cells is heterogeneous:down-regulated in a subgroup of noninflammatory pa-tients and up-regulated in a subgroup of inflammatorypatients (47, 48). The macrophage phenotype of patientswith MCI is also heterogeneous and either highly proin-flammatory M1 or very low inflammatory M2 (49). Thus,v-3 therapy should lead to an intermediate proresolutionM1-M2 phenotype, as shown in a pilot study (49).

IMMUNE THERAPIES BY RECEPTORANTAGONISTS, v-3 AND v-3–DERIVEDSPECIALIZED PRORESOLVING MEDIATORS,AND EPOXY FATTY ACIDS

Immuneapproaches in animalmodels, suchas chemokinereceptor CCR2 antagonists, purinergic receptor P2Y6 an-tagonists, or a CX3CR1 agonist (31), target single genes inCNS myeloid cells and do not consider the heterogeneityof inflammation or immune exhaustionwith deregulationof many genes and pathways in amyotrophic lateral scle-rosis (50, 51) and AD (49). Remarkably, lipid mediators

2 Vol. 31 August 2017 FIALA ET AL.The FASEB Journal x www.fasebj.org Vol.,  No. , pp:, April, 2017The FASEB Journal. 169.237.169.81 to IP www.fasebj.orgDownloaded from

Page 3: Modulation of innate immunity of patients with Alzheimer’s ... · THE JOURNAL † REVIEW † Modulation of innate immunity of patients with Alzheimer’s disease by omega-3 fatty

from v-3 regulate the activation of a multitude of genesand pathways that are related to inflammation and reso-lution in vitro and in vivo (52) and provide a different ap-proach to AD therapy by multitargeting. Inflammation isnormally terminated by an active process called resolu-tion, which limits the entry of immune cells, increases theclearance of apoptotic cells by macrophage phagocytosis,and enables tissue restoration (53). Incomplete resolutionleads to unabated inflammation, which is an underlyingcause of many chronic diseases (54–56).

During the resolution of acute inflammation, host-protective mediators are biosynthesized from poly-unsaturated fatty acids (PUFA) via up-regulation of the keybiosynthetic enzyme15-lipoxygenase type 1 in leukocytes(57). This promotes the conversion of v-6 arachidonicacid to lipoxin A4 (LXA4) and of v-3 DHA to D-series

resolvins and protectins/neuroprotectins (57). DHA isalso converted in macrophages via 14-lipoxygenation tomaresins (macrophagemediators in resolving inflammation).The first identified member of this family was maresin 1(MaR1; 7R,14S-dihydroxy-docosa-4Z,8E,10E,12Z,16Z,19Z-hexaenoic acid). Specific members of these families ofmediators from the DHA metabolome are namedD-series resolvins [resolvin D1 (RvD1) to -6], pro-tectins (including protectin D1-neuroprotectin D1),and maresins (MaR1 and -2) and E-series resolvins(RvE1 and -2) (58). These mediators limit neutro-phil recruitment and potently stimulate macrophagephagocytosis of apoptotic cells in a stereospecificmanner. They also regulate both inflammation andchemotherapy-induced pain by inhibiting transientreceptor potential vanilloid 1 currents inneurons (59).A

THE AD Brain

Hypoglycemia, hypoxemia,dyslipidemia

Mitochondrial energy disruptionVirusesPrions

Neurotoxins

Aβ accumulationtau phosphorylation

Invading macrophages:Classical (M1)

Alternative (M2)Intermediate (M1-M2)

Disease progressionω-3 supplementation

MΦVery high M1 signature - decreased Aβ phagocytosis and disease progressionVery low M2 signature - decreased Aβ phagocytosis and disease progressionIntermediate M1-M2 signature - increased Aβ phagocytosis, decreased apoptosis

MΦ invasion across BBB

Z0-1 Disruption

Macrophage

mϕ in an Aβ plaqueMΦ Green with Aβ

Plaque (Red)

Aβ Green

MΦ (Yellow)

apoptotic perlvascular macrophages

leaky vessels

Fibrimogen

A B

a

b

c

d

a

b

c

d

Figure 1. A) Immunopathology of Alzheimer’s disease: pathogenic stimuli (a); Ab accumulation and tau phosphorylation (b);M1, -2 or M1-M2 macrophage phenotypes before modulation (c); v-3 do or do not modulate the phenotype (d); M1, -2 or M1-M2phenotypes after modulation (e). B) Immunochemistry of AD brain: macrophage invasion across BBB (a); macrophage invasionof Ab plaque (b); perivascular macrophages around congophilic vessels (c); leaky cerebral vessel (d) (17).

MODULATION OF IMMUNITY IN ALZHEIMER’S DISEASE 3 Vol.,  No. , pp:, April, 2017The FASEB Journal. 169.237.169.81 to IP www.fasebj.orgDownloaded from

Page 4: Modulation of innate immunity of patients with Alzheimer’s ... · THE JOURNAL † REVIEW † Modulation of innate immunity of patients with Alzheimer’s disease by omega-3 fatty

novel family of macrophage-derived proresolving media-tors, called Maresins Conjugates in Tissue Regeneration,carry potent organ-protective, tissue-regenerative, and pro-resolving actions (60). In addition, protectin and resolvinsulfido-conjugates were recently identified as novelresolution mediators that control regeneration (60, 61).

Specialized proresolving mediators (SPMs) bind to theirreceptorson leukocytesandregulateanti-inflammatoryandproresolving actions. LXA4 inhibits leukocyte trafficking invivo by activating the LXA4 receptor denoted ALX/FPR2.RvD1 binds to the ALX/FPR2 receptor and another GPCRcalledGPR32, but RvD1 does not activate nuclear receptorsretinoid X receptor (RXR)-a and peroxisome proliferator-activated receptor (PPAR)-a, -d, and -g (62). RvD1 inhibitsneutrophil migration and promotes their phagocytosis bymacrophages (62). Aspirin-triggered RvD1 (AT-RvD1) in-hibits the release of IL-6 and protects the brain after surg-ical injury from neuroinflammation, synaptic dysfunction,and cognitive decline (63). RvD2 binds to GPCR GPR18and mediates bacterial phagocytosis and organ protection(64). RvE1 binds to the receptor on macrophages calledChemR23 (65), also named human chemokine-like receptor1. 13S,14S-Epoxide-maresin intermediate stimulates M1 toM2 phenotype switch (66). PD1 inhibits inflammatory cy-tokines, COX-2, and NF-kB in the CNS (67).

These SPMs are active in animal models of differentinfectious, metabolic, ischemic, arthritic, and oncogenicpathologies, such asRvD2 inmicrobial sepsis (68), RvE1 inperiodontal disease (69), PD1 in ocular herpes simplex(70), LXA4 in liver and renal fibrosis related to obesity (71),LXA4 in cerebral ischemia (72), LXA4 in brain amyloidosis(73), RvD1 in arthritis (74), and resolvins in cancer (75).Emerging studies in human participants show positivetherapeutic effects of v-3 supplementation on clot re-sorption bymacrophages of patients with coronary arterydisease (76) and on phagocytosis of Ab mentioned above(48).

v-3 PUFAs are also a source for the formation of epoxyfatty acids (EpFAs), epoxydocosapentaenoic acids fromDHA, and EETs from EPA (77) (Table 1). EpFAs areantinociceptive in several models of pain, and, like resol-vins, they dampen prostaglandin- and cytokine-driveninflammation (7, 78, 79).Of interest,v-6PUFAarachidonicacid is also a precursor to anti-inflammatory lipid media-tors that are formed in different branches of the arachi-donic acid cascade. The biologic activities of LXA4 formedby lipoxygenaseand thoseofEETsgeneratedbycytochromeP450 enzymes oppose those of prostaglandin metabolites

formedby cyclooxygenase activity on the arachidonic acidsubstrate (80). EETs are anti-inflammatory and anti-nociceptive, whereas prostaglandins and leukotrienesare inflammatory and nociceptive (81–83); however,prostaglandins are also involved in lipid-mediator classswitching in which these classic initiators of acute in-flammation switch to produce SPMs (84).

An enzyme downstream of cytochrome P450 activity,soluble epoxide hydrolase (sEH), is a master regulatingenzyme of the different EpFA metabolite classes. In-hibition of sEH stabilizes EpFAs, inhibits inflammation,and reduces pain (85). The action of EpFAs has multiplemechanisms, including interaction with the cAMP path-way (86), in addition to reduction of endoplasmic re-ticulum stress (87). In the brain, sEH inhibition and anincrease in EpFAs have reduced damage from cerebralischemia inboth aneuroprotectiveandvascularprotectivemanner (88).Of importance, EETs exert this protective rolevia glia in addition to vascular cells (89). There is evidencefrom multiple sources that high levels of sEH and a de-crease in epoxide/diol ratios is associated with vasculardementia (90) and with several psychiatric disorders, in-cluding depression (91) and schizophrenia (92).

SPMS IN THE AD BRAIN

In thebrain,DHAisaprecursor to theneuroprotectiveandanti-inflammatorymediator, PD1 (93), which is decreasedin the AD brain (94) and reduces Ab-induced neurotox-icity (95). Moreover, it has recently been shown that LXA4

wasdecreased in theADbrain andCSFand thatLXA4andRvD1 levels in the CSF positively correlate with mini-mental state examination (MMSE) scores (8), therebysuggesting a connectionwith cognitive function inAD. Inlinewith these findings, Zhu and colleagues (55) recentlyreported lower levels of MaR1, PD1, and RvD5 in theentorhinal cortex of patients with AD, thereby providingmore evidence of a disturbed resolution pathway in AD.Addition of these SPMs exerted neuroprotective activityand boosted Ab uptake by microglia, which illustratedthe feasibility of use of SPMs in a clinical setting. Indeed,supplementation with AT-RvD1 preventedmemory lossin an animal model of surgery-induced cognitive decline(63), and AT-LXA4 improved cognition in a transgenicmouse model of AD (96).

TABLE 1. PUFAs and proresolution lipid mediators

Precursor Lipid mediator Effects of lipid mediator Reference

v-3 fatty acidDHA D-series resolvins: RvD1–D6, Mar1, -2 Prophagocytic and anti-inflammatory 46EPA E-series resolvins: RvE1–E3DHA Epoxydocosapentaenoic acids Anti-inflammatory and antinociceptive 63EPA EEQ Anti-inflammatory and antinociceptive 7

v-6 fatty acidArachidonic acid LXA4 Anti-inflammatory and prophagocytic 160

EETs Anti-inflammatory and antinociceptive 7

4 Vol. 31 August 2017 FIALA ET AL.The FASEB Journal x www.fasebj.org Vol.,  No. , pp:, April, 2017The FASEB Journal. 169.237.169.81 to IP www.fasebj.orgDownloaded from

Page 5: Modulation of innate immunity of patients with Alzheimer’s ... · THE JOURNAL † REVIEW † Modulation of innate immunity of patients with Alzheimer’s disease by omega-3 fatty

MACROPHAGE PHENOTYPES IN PATIENTS WITHAD AND MCI AND POLARIZATION BY v-3AND SPMS

Blood-bornemonocyte-derivedmacrophages have criticalimportance in various neurodegenerative diseases, asthese cells cross the BBB and produce specific immunepathologies in HIV-1 encephalitis (20), AD (17), andamyotrophic lateral sclerosis (97). Macrophages are bi-functional in Ab phagocytosis: in healthy individuals,they are effective in phagocytosis and the degradation ofAb, whereas in patientswithMCI andAD,macrophagesare defective in both functions (98).

Regulationof themacrophagephenotype is at the coreoftherapy for a variety of disorders, including atherosclerosis,allergy, infection, tissuerepair, obesity, emphysema, cancer,and neurodegenerative diseases (99). The extremes of amacrophage phenotype continuum in model systems aredescribed as M1 proinflammatory and M2 proresolvingand wound-healing macrophages, but in vivo intermediateand unique macrophage phenotypes are frequently found(100, 101). M1 macrophages produce cytotoxic and in-flammatory cytokines (TNF-a and IL-12), chemokines(CXCL9, -10, -11), andNOS2.M2macrophages up-regulatethe mannose receptor (Mrc1) and chitinase 3-like3 (Chi3l3).Macrophagepolarization isactivated in the followingways:1) toM1 phenotype by IFN-a/b or IFN-g signaling via IFNregulatory factor-5 (IRF5) and signal transducer and acti-vator of transcription 1 (STAT1), or alternatively by LPS viaTLR4andNF-kB signaling; 2) toM2phenotype by IL-4 andIL-13 signaling via STAT6, PPAR-g, and PPAR-d (102); IL-4induces Jmjd3-IRF4 axis to inhibit IRF5-mediated M1 po-larization (103); 3) by epigenetic changes, including thosevia a histone demethylase (104); and 4) bymiR-155 (105). Inaddition, T cells influence macrophage polarization: Th1stimulateM1 type and regulatory T cells stimulateM2 typephenotypes (106).

Defects in AD macrophage phagocytosis seem to berelated to defective protein glycosylation as a result ofdown-regulation of the enzyme, b-1,4-mannosyl-glyco-protein4-b-N-acetylglucosaminyltransferase (GlcNAc).Thisenzyme is up-regulated during Ab phagocytosis in nor-mal, but not AD, monocytes (107). Low expression ofGlcNAc is associated with poor prognosis (108). As theinhibition ofN-glycosylation decreases anti-inflammatoryand proresolution M2 macrophage polarization (109), re-duced transcription ofGlcNAcmaybe an important defectin patients with AD. b-Site amyloid precursor proteincleaving enzyme-1 is modified by bisecting GlcNAc in thebrain of patients with AD, which possibly leads to greaterb-site amyloid precursor protein cleaving enzyme-1 deg-radation and, thus, increased Ab production, as observedin a mouse model (110).

Macrophage polarization toward the proresolving M2type by SPM has a favorable effect in many disorders. v-3protect against obesity-associatedmetabolic disorders andcardiovascular disease via modulation of eicosanoid pro-duction and stimulation of anti-inflammatory action.RvD1 inhibits macrophage accumulation in adipose tis-sues and increases the proportion of alternatively acti-vated macrophages (111, 112). Resolvins, maresins, and

13S,14S-epoxy-maresin shift macrophage phenotype to aless inflammatory type (62, 113). SPMs polarize inflamma-toryM1macrophage type in amouse-air-pouchmodel to aproresolution M2 type (114). M2 macrophages have ele-vated levels of proresolving lipid mediators, includingmaresin, and decreased prostaglandins and leukotrienes inrelation to M1 macrophages (115). In addition, M2 macro-phages specificallyproduce the recently identifiedprotectinconjugate in tissue regeneration 1, which seems to be apotent novel inducer of macrophage phagocytosis (116).SPMs alsomodulate themetabolism ofmacrophages (117).

RvD1 interacts with GPCR GPR32 (62), whereas fattyacids employ PPAR-a (118–120). v-3 DHA induces M2macrophage polarization and efferocytosis via PPAR-g(121). In mouse models of AD and in vitro microglia andastrocytes, DHA has anti-inflammatory and proresolvingeffects via RXR and PPAR-g (122).

STIMULATION OF INNATE IMMUNITY BYNATURAL SUBSTANCES

In 2016, the monumental action against AD includes thedevelopment of nontoxic therapies with Abs and naturalsubstances. This approachhasgained traction after clinicalfailures and amyloid-related imaging abnormalities of AbAbs (123), which also increased cortical hyperactivity in amouse model (124). The next generations of the vaccineand Abs in development are designed to clear Ab by ac-tively or passively induced AbAbs, not by improving thefunction ofmacrophages and/ormicroglia, andmayhavevarious adverse effects on chronic administration (125).Recent Ab Ab approaches include gantenerumab (126)and aducanumab (127). Aducanumab reduced brain Aband putatively slowed clinical decline but had Ab-relatedimaging abnormalities.

Natural substances havebeenused for centuries to boostthe immune system with manifest results, such as that ob-served with curcuminoids (128) and vitamin D3 (129). De-spite extensive anecdotal experience, clinical investigationof natural substances inMCI andAD is difficult because ofthe chronic nature of these diseases that requires largeprospective studies. A large compendium of nutritionalsupplements for dementia (130) includes a cornucopia ofmicronutrients, minerals, vitamins (1,25D3 and vitamin Bcomplex), v-3 fatty acids DHA and EPA, flavonoids (e.g.,resveratrol), polyphenols (e.g., curcuminoids), and alka-loids (caffeine, ashwagandha), as well as nutraceuticals,suchasherbs,diets, andprobiotics.Currently, the followingsubstances have a potential for prevention of MCI pro-gression via increased Ab clearance on the basis of clinicalor experimental experience (131): v-3 fatty acids (48), vita-min D3 (132), vitamin B complex (133), and curcuminoids(35, 134–136).

MECHANISMS OF SPMS, 1,25D3, AND OTHERMEDIATORS THAT ENHANCE Ab PHAGOCYTOSIS

v-3 and 1,25D3 are the best-understood natural im-mune therapeutics for human use in patients with

MODULATION OF IMMUNITY IN ALZHEIMER’S DISEASE 5 Vol.,  No. , pp:, April, 2017The FASEB Journal. 169.237.169.81 to IP www.fasebj.orgDownloaded from

Page 6: Modulation of innate immunity of patients with Alzheimer’s ... · THE JOURNAL † REVIEW † Modulation of innate immunity of patients with Alzheimer’s disease by omega-3 fatty

MCI, but their effects in patients are heterogeneous.The effects of v-3 and 1,25D3 are different from eachother and do not completely normalize the AD tran-scriptome (6). Both mediators up-regulated IL-1 innoninflammatory patients and down-regulated IL-1and IL-6 in inflammatory patients. In general, 1,25D3had more pronounced effects on normalization ofchemokine and cytokine transcription than did RvD1.1,25D3 down-regulated IL-1R1, CCL23, -2, and CD40in both groups of patients. RvD1 profoundly down-regulated NF-kB, IL-1b, CCL2, -4, and -24 in in-flammatory patients, but up-regulated IL-1a and IL-1bin noninflammatory patients. 1,25D3 forms heterodimerswith RXR and stimulates Ab phagocytosis via non-genomic signaling by increasing intracellular calciumand potentiating MEK1/2 signaling, which leads to theopening of the ClC3 channel, as well as via nuclear sig-naling (47).

Other chemicals that enhance phagocytosis in-clude bexarotene and resveratrol. Bexarotene is anRXR agonist that activates the PPAR-g and RXR het-erodimer. Bexarotene potentiates phagocytosis of Abin an animal model (137, 138), but this has been onlyincompletely reproduced in other laboratories (139).Resveratrol altered certain CSF biomarkers (Ab1–40)without consistent cognitive changes in a controlledclinical trial (140). Resveratrol has a broad spectrumof activities against Ab and t neuropathologies, in-cluding attenuation of inflammatory mechanismsand increasing intracellular Ab clearance, in part byactivating autophagy and proteolysis viamodulationof the AMPK/sirtuin1/PPAR-g coactivator1-a sig-naling network (141). PPAR-g coactivator1-a is atranscriptional coactivator that regulates the genesthat are involved in energy metabolism and mito-chondrial biogenesis and function and induces po-larization of macrophages toward M2 via STAT6/PPAR-g (142).

OBSERVATIONAL STUDIES AND CLINICALTRIALS OF v-3: COGNITIVE, IMMUNE, ANDMOLECULAR EFFECTS

Retrospective epidemiologic studies in various pop-ulations have shown positive cognitive results in relationto optimal DHA plasma levels or a high consumption offish in 14 of 17 studies (143). In a controlled study of v-3supplementation, patients with MCI with MMSE score.27 reduced cognitive decline compared with placebo(144). v-3 supplementation in a double-blind study im-proved memory functions in healthy older adults (145).Recently, 3 trials (144–147)were analyzed according to theCochrane Handbook for Systematic Reviews of Interven-tions and found no evidence for the efficacy of v-3 sup-plements in the treatment of mild to moderate AD (148).Seafood consumption ($1 meal/wk) was significantlycorrelated with less AD pathology, but this effect wasobserved only among apolipoprotein E (APOE) e4 carriers(149). The choice of fish-derived DHA vs. algal DHA hasnot been considered an issue, but the 2 largest clinical trials

of DHA supplementation had different results: fish-derived DHA showed the slowing of decline in patientswith early MCI, whereas algal DHA was not differentfrom placebo.

A recent observational study of supplementation bythe v-3 drink, Smartfish (Smartfish, Oslo, Norway),analyzed the immune and molecular effects of v-3supplementation in patients with MCI. This drink is awell-characterized, stabilized emulsion of v-3 lipids,antioxidants, vitamin D3, whey protein, and resvera-trol. This small study showed significant enhancementof Ab phagocytosis after only 1 mo in parallel withcognitive stabilization (48). Immune improvementwas marred in some patients by lack of daily compli-ance, infections, diabetes mellitus, cancer, and sur-geries. RvD1 increased in macrophages in 80% ofpatients with MCI. v-3 regulated the inflammatoryactivation of peripheral blood mononuclear cells—either too high or too low at baseline—into a middlegreen zone (48). This study has been recently extendedto 14.7 mo (average follow-up) (49). The results haveshown that macrophage M1 or -2 type at baseline wasmodulated in v-3–supplemented APOEe3/e3 patientsto an intermediate type M1-M2 with improved Abphagocytosis, whereas macrophages of APOEe3/e4patients were modulated in an irregular fashion. Ofimportance, MMSE score significantly increased inAPOEe3/e3 patients (by 2.2 points/yr) vs. no increasein APOEe3/e4 patients (0 points/yr) (49).

DESIGN OF AN IDEAL CLINICAL TRIAL OFv-3 SUPPLEMENTATION

It is clear that MCI and AD involve the deregulation ofmany pathways and thus requires combination therapy,as suggested by the glimmers of success with this ap-proach (150). Effective therapy should be based on v-3 ofthe highest quality, protected against oxidation [v-3 areoxidized in 50%of commercial supplements (151)]. It is notclearwhether there are advantages of fish-derivedvs. algalv-3 and whether DHA, EPA, or both are needed. Wheyprotein is necessary for emulsification and nutrient de-livery (152). Additional natural supplements with centraland peripheral immune effects, such as resveratrol (153),vitamin D3 (154), vitamin B complex (folic acid, vitaminsB6 and B12) (155), and curcumin (35), should be evaluatedfor inclusion in the supplement. The design should berandomized, placebo-controlled, anddouble-blind,with asample size of adequate power. Participants should beobserved in 2-mo intervals and comorbidities, such asfractures, infections, and cancer, whichmay interfere withthe immune response, must be analyzed. As it is impos-sible to know the genotype in advance of trial enrollment,results must be analyzed according to the APOEe3/e3 vs.e3/e4 genotype. Use of over-the-counter supplementsshould be discontinued. According to the results of ourrecent trial of v-3 (49), we estimate that in APOEe3/e3patientswithMCI, the effectof Smartfish supplementationonMMSE could be 60–70% greater than that of a placebo.This is more likely to be achieved if patients follow

6 Vol. 31 August 2017 FIALA ET AL.The FASEB Journal x www.fasebj.org Vol.,  No. , pp:, April, 2017The FASEB Journal. 169.237.169.81 to IP www.fasebj.orgDownloaded from

Page 7: Modulation of innate immunity of patients with Alzheimer’s ... · THE JOURNAL † REVIEW † Modulation of innate immunity of patients with Alzheimer’s disease by omega-3 fatty

recommendations for physical and mental exercise, ade-quate sleep, and eating a diet low in saturated fat and highin fish (156). Assuming 60% effect, we calculated thesample size in a study with a 2-yr follow-up (with 80%power, a = 0.05) as 30 patients on v-3 preparation and 30controls on a placebo in an ideal situation with identicalproportion of APOEe3/e3 vs. e3/e4 genotype subjects;however, the APOE genotype distribution cannot be esti-mated in advance in small samples. Therefore, the numberof the participants in the trial should be at least 2 or 3 timeshigher. Trials of mAbs against Ab required 2000 patients(157, 158) and still failed; however, even a small-size(100–200 participants) trial of v-3 preparation will de-finitively produce a wealth of data on immune responsesthatmay explainwhy certain patients respond cognitivelybetter than others.

CONCLUSIONS

A retrospective (143) and a prospective study (144) dem-onstrated cognitive benefits with v-3 supplementation.Our observational study with the v-3 drink suggests thatthe beneficial effects are associated with macrophage po-larization to M1-M2 type, particularly in APOe3/e3 pa-tients (49). Conversely, 2 large prospective studies seem todisclaim the benefits of v-3 because oral supplementationwith algal DHA (but without antioxidants and othercomponents) had no effect on the cognitive function inpatients who were at risk of macular degeneration (159)and in those with AD (146). The benefits of v-3 need to beconfirmed in a randomized controlled trial that will ana-lyze not only the overall cognitive results but also baselinecognitive and health status, APOE type, compliance (i.e., areliable caregiver), intercurrent inflammatory and immu-nosuppressiveconditions, andcorrelationof cognitiveandimmune results during the trial.

ACKNOWLEDGMENTS

Studies of innate immunity in AD and MCI were supportedby a grant from the Alzheimer’s Association and a donationfrom Smartfish AS. Smartfish AS supplied the drink to patientswho requested it, but was not involved in the design, analysis,interpretation of the data, or the decision to submit themanuscript for publication. M.F. received paid travel tomeetings and honoraria from Smartfish AS. G.K. receivedsupport from the Dutch MS Research Foundation (14-878MS). B.H. and K.W. were supported by U.S. National Institutesof Health, National Institute of Environmental Health SciencesGrant R01-ES002710, and K.W. by Grant 5T32L086350-08. Theauthors thank J. Gornbein (Department of Biomathematics,University of California Los Angeles) for calculating thesample size of the v-3 study.

AUTHOR CONTRIBUTIONS

M.FialaandM.Pellegrini reviewedv-3 studies inmacrophagesand human patients; G. Kooij reviewed specialized pro-resolving mediators; and K. Wagner and B. Hummockreviewed epoxy fatty acids.

REFERENCES

1. Petersen,R.C.,Doody, R., Kurz,A.,Mohs,R.C.,Morris, J. C., Rabins,P. V., Ritchie, K., Rossor, M., Thal, L., and Winblad, B. (2001)Current concepts in mild cognitive impairment. Arch. Neurol. 58,1985–1992

2. Duara, R., Loewenstein, D. A., Greig, M. T., Potter, E., Barker, W.,Raj, A., Schinka, J., Borenstein,A., Schoenberg,M.,Wu, Y., Banko, J.,and Potter, H. (2011) Pre-MCI and MCI: neuropsychological, clin-ical, and imaging features and progression rates. Am. J. Geriatr. Psy-chiatry 19, 951–960

3. Roberts, R. O., Knopman,D. S., Mielke,M.M., Cha, R. H., Pankratz,V. S., Christianson, T. J., Geda, Y. E., Boeve, B. F., Ivnik, R. J.,Tangalos, E. G., Rocca,W.A., and Petersen, R. C. (2014)Higher riskof progression to dementia in mild cognitive impairment cases whorevert to normal. Neurology 82, 317–325

4. Hardy, J., and Selkoe, D. J. (2002) The amyloid hypothesis ofAlzheimer’s disease: progress and problems on the road totherapeutics. Science 297, 353–356

5. Serhan, C. N. (2014) Pro-resolving lipid mediators are leads forresolution physiology. Nature 510, 92–101

6. Mizwicki, M. T., Liu, G., Fiala, M., Magpantay, L., Sayre, J., Siani, A.,Mahanian, M., Weitzman, R., Hayden, E. Y., Rosenthal, M. J.,Nemere, I., Ringman, J., and Teplow, D. B. (2013) 1a,25-dihydroxyvitamin D3 and resolvin D1 retune the balance betweenamyloid-b phagocytosis and inflammation in Alzheimer’s diseasepatients. J. Alzheimers Dis. 34, 155–170

7. Wagner, K., Vito, S., Inceoglu, B., and Hammock, B. D. (2014) Therole of long chain fatty acids and their epoxide metabolites innociceptive signaling.ProstaglandinsOtherLipidMediat.113-115, 2–12

8. Wang, X., Zhu, M., Hjorth, E., Cortes-Toro, V., Eyjolfsdottir, H.,Graff, C., Nennesmo, I., Palmblad, J., Eriksdotter, M., Sambamurti,K., Fitzgerald, J.M., Serhan,C.N.,Granholm,A.C., andSchultzberg,M. (2015) Resolution of inflammation is altered in Alzheimer’sdisease. Alzheimers Dement. 11, 40–50.e1-2

9. Simard, A. R., Soulet, D., Gowing, G., Julien, J. P., and Rivest, S.(2006) Bone marrow-derived microglia play a critical role inrestricting senile plaque formation in Alzheimer’s disease. Neuron49, 489–502

10. Rogers, J., and Lue, L. F. (2001) Microglial chemotaxis, activation,and phagocytosis of amyloid beta-peptide as linked phenomena inAlzheimer’s disease. Neurochem. Int. 39, 333–340

11. Akiyama, H., Arai, T., Kondo, H., Tanno, E., Haga, C., and Ikeda, K.(2000) Cell mediators of inflammation in the Alzheimer diseasebrain. Alzheimer Dis. Assoc. Disord. 14, S47–S53

12. Fiala, M., Zhang, L., Gan, X., Sherry, B., Taub, D., Graves, M. C.,Hama, S., Way, D., Weinand, M., Witte, M., Lorton, D., Kuo, Y. M.,and Roher, A. E. (1998) Amyloid-beta induces chemokine secretionand monocyte migration across a human blood–brain barriermodel.Mol. Med. 4, 480–489

13. El Khoury, J., Toft, M., Hickman, S. E., Means, T. K., Terada, K.,Geula, C., and Luster, A. D. (2007) Ccr2 deficiency impairsmicroglial accumulation and accelerates progression of Alzheimer-like disease. Nat. Med. 13, 432–438

14. Atkins, H. L., and Freedman, M. S. (2013) Hematopoietic stem celltherapy for multiple sclerosis: top 10 lessons learned.Neurotherapeutics 10, 68–76

15. Ginhoux, F., Greter, M., Leboeuf, M., Nandi, S., See, P., Gokhan, S.,Mehler, M. F., Conway, S. J., Ng, L. G., Stanley, E. R., Samokhvalov,I.M., andMerad,M. (2010) Fatemapping analysis reveals that adultmicroglia derive from primitive macrophages. Science 330, 841–845

16. Wisniewski,H.M.,Barcikowska,M., andKida,E. (1991)Phagocytosisof beta/A4 amyloid fibrils of the neuritic neocortical plaques. ActaNeuropathol. 81, 588–590

17. Fiala, M., Liu, Q. N., Sayre, J., Pop, V., Brahmandam, V., Graves,M. C., and Vinters, H. V. (2002) Cyclooxygenase-2-positive macro-phages infiltrate the Alzheimer’s disease brain and damage theblood-brain barrier. Eur. J. Clin. Invest. 32, 360–371

18. Goldmann, T., Wieghofer, P., Jordão, M. J., Prutek, F., Hagemeyer,N., Frenzel, K., Amann, L., Staszewski, O., Kierdorf, K., Krueger, M.,Locatelli,G.,Hochgerner,H., Zeiser,R., Epelman,S.,Geissmann,F.,Priller, J., Rossi, F. M., Bechmann, I., Kerschensteiner, M.,Linnarsson, S., Jung, S., and Prinz, M. (2016) Origin, fate anddynamics of macrophages at central nervous system interfaces.Nat.Immunol. 17, 797–805

19. Hagemeyer, N., Kierdorf, K., Frenzel, K., Xue, J., Ringelhan, M.,Abdullah, Z., Godin, I., Wieghofer, P., Costa Jordão, M. J., Ulas, T.,

MODULATION OF IMMUNITY IN ALZHEIMER’S DISEASE 7 Vol.,  No. , pp:, April, 2017The FASEB Journal. 169.237.169.81 to IP www.fasebj.orgDownloaded from

Page 8: Modulation of innate immunity of patients with Alzheimer’s ... · THE JOURNAL † REVIEW † Modulation of innate immunity of patients with Alzheimer’s disease by omega-3 fatty

Yorgancioglu, G., Rosenbauer, F., Knolle, P. A., Heikenwalder, M.,Schultze, J. L., and Prinz, M. (2016) Transcriptome-based profilingof yolk sac-derived macrophages reveals a role for Irf8 in macro-phage maturation. EMBO J. 35, 1730–1744

20. Persidsky, Y., Ghorpade, A., Rasmussen, J., Limoges, J., Liu,X. J., Stins, M., Fiala, M., Way, D., Kim, K. S., Witte, M. H.,Weinand, M., Carhart, L., and Gendelman, H. E. (1999)Microglial and astrocyte chemokines regulate monocytemigration through the blood-brain barrier in human immu-nodeficiency virus-1 encephalitis. Am. J. Pathol. 155, 1599–1611

21. Ajami, B., Bennett, J. L., Krieger, C., Tetzlaff, W., and Rossi, F. M.(2007) Local self-renewal can sustain CNS microglia maintenanceand function throughout adult life. Nat. Neurosci. 10, 1538–1543

22. Mildner, A., Schmidt, H., Nitsche, M., Merkler, D., Hanisch, U. K.,Mack, M., Heikenwalder, M., Bruck, W., Priller, J., and Prinz, M.(2007) Microglia in the adult brain arise from Ly-6ChiCCR2+

monocytes only under defined host conditions. Nat. Neurosci. 10,1544–1553

24. ElAli, A., and Rivest, S. (2016) Microglia in Alzheimer’s disease: amultifaceted relationship. Brain Behav. Immun. 55, 138–150

23. Hawkes, C. A., and McLaurin, J. (2009) Selective targeting ofperivascular macrophages for clearance of beta-amyloid in cerebralamyloid angiopathy. Proc. Natl. Acad. Sci. USA 106, 1261–1266

25. Paresce, D. M., Chung, H., and Maxfield, F. R. (1997) Slowdegradation of aggregates of the Alzheimer’s disease amyloid beta-protein by microglial cells. J. Biol. Chem. 272, 29390–29397

26. Mildner, A., Schlevogt, B., Kierdorf, K., Bottcher, C., Erny, D.,Kummer,M. P.,Quinn,M., Bruck,W., Bechmann, I.,Heneka,M.T.,Priller, J., and Prinz, M. (2011) Distinct and non-redundant rolesof microglia and myeloid subsets in mouse models of Alzheimer’sdisease. J. Neurosci. 31, 11159–11171

27. Baruch, K., Deczkowska, A., Rosenzweig, N., Tsitsou-Kampeli, A.,Sharif, A. M., Matcovitch-Natan, O., Kertser, A., David, E., Amit, I.,and Schwartz, M. (2016) PD-1 immune checkpoint blockade re-duces pathology and improves memory in mouse models of Alz-heimer’s disease. Nat. Med. 22, 135–137

28. Baruch,K.,Rosenzweig,N.,Kertser,A.,Deczkowska,A., Sharif,A.M.,Spinrad, A., Tsitsou-Kampeli, A., Sarel, A., Cahalon, L., andSchwartz, M. (2015) Breaking immune tolerance by targetingFoxp3+ regulatory T cells mitigates Alzheimer’s disease pathology.Nat. Commun. 6, 7967

29. Bennett, M. L., Bennett, F. C., Liddelow, S. A., Ajami, B., Zamanian,J. L., Fernhoff,N. B.,Mulinyawe, S. B., Bohlen, C. J., Adil, A., Tucker,A., Weissman, I. L., Chang, E. F., Li, G., Grant, G. A.,Hayden Gephart, M. G., and Barres, B. A. (2016) New tools forstudying microglia in the mouse and human CNS. Proc. Natl. Acad.Sci. USA 113, E1738–E1746

30. Meyer-Luehmann, M., and Prinz, M. (2015) Myeloid cells inAlzheimer’s disease: culprits, victims or innocent bystanders?Trends Neurosci. 38, 659–668

31. Biber, K., Moller, T., Boddeke, E., and Prinz, M. (2016) Centralnervous system myeloid cells as drug targets: current status andtranslational challenges. Nat. Rev. Drug Discov. 15, 110–124

32. Schneider, L. S., Mangialasche, F., Andreasen, N., Feldman, H.,Giacobini, E., Jones, R., Mantua, V., Mecocci, P., Pani, L., Winblad,B., and Kivipelto, M. (2014) Clinical trials and late-stage drug de-velopment for Alzheimer’s disease: an appraisal from 1984 to 2014.J. Intern. Med. 275, 251–283

33. Prokop, S., Miller, K. R., Drost, N., Handrick, S., Mathur, V.,Luo, J., Wegner, A., Wyss-Coray, T., and Heppner, F. L. (2015)Impact of peripheral myeloid cells on amyloid-b pathology inAlzheimer’s disease-like mice. J. Exp. Med. 212, 1811–1818

34. Jay, T., Lamb, B., and Landreth, G. (2015) Peripheral macrophagesnot ADept at amyloid clearance. J. Exp. Med. 212, 1758

35. Fiala, M., Liu, P. T., Espinosa-Jeffrey, A., Rosenthal, M. J.,Bernard, G., Ringman, J. M., Sayre, J., Zhang, L., Zaghi, J.,Dejbakhsh, S., Chiang, B., Hui, J., Mahanian, M., Baghaee, A.,Hong, P., and Cashman, J. (2007) Innate immunity andtranscription of MGAT-III and Toll-like receptors in Alz-heimer’s disease patients are improved by bisdemethoxy-curcumin. Proc. Natl. Acad. Sci. USA 104, 12849–12854

36. Mizwicki, M. T., Bula, C. M., Mahinthichaichan, P., Henry,H. L., Ishizuka, S., and Norman, A. W. (2009) On themechanism underlying (23S)-25-dehydro-1alpha(OH)-vitaminD3-26,23-lactone antagonism of hVDRwt gene activation and itsswitch to a superagonist. J. Biol. Chem. 284, 36292–36301

37. Seok, J.,Warren,H. S., Cuenca, A.G.,Mindrinos,M.N., Baker,H.V.,Xu, W., Richards, D. R., McDonald-Smith, G. P., Gao, H., Hennessy,L., Finnerty, C. C., Lopez, C.M.,Honari, S.,Moore, E. E.,Minei, J. P.,Cuschieri, J., Bankey, P. E., Johnson, J. L., Sperry, J., Nathens, A. B.,Billiar, T. R., West,M. A., Jeschke, M. G., Klein,M. B., Gamelli, R. L.,Gibran, N. S., Brownstein, B. H., Miller-Graziano, C., Calvano, S. E.,Mason, P. H., Cobb, J. P., Rahme, L. G., Lowry, S. F., Maier, R. V.,Moldawer, L. L., Herndon, D. N., Davis, R. W., Xiao, W., andTompkins, R. G.; Inflammation andHost Response to Injury, LargeScale Collaborative Research Program. (2013) Genomic responsesin mousemodels poorly mimic human inflammatory diseases. Proc.Natl. Acad. Sci. U. S. A. 110, 3507-3512

38. Colton, C. A., Wilson, J. G., Everhart, A., Wilcock, D.M., Puolivali, J.,Heikkinen,T.,Oksman, J., Jaaskelainen,O., Lehtimaki, K., Laitinen,T., Vartiainen, N., and Vitek, M. P. (2014) mNos2 deletion andhuman NOS2 replacement in Alzheimer disease models.J. Neuropathol. Exp. Neurol. 73, 752–769

39. Zaghi, J., Goldenson, B., Inayathullah,M., Lossinsky, A. S.,Masoumi,A., Avagyan,H.,Mahanian,M., Bernas,M.,Weinand,M., Rosenthal,M. J., Espinosa-Jeffrey, A., de Vellis, J., Teplow, D. B., and Fiala, M.(2009) Alzheimer disease macrophages shuttle amyloid-beta fromneurons to vessels, contributing to amyloid angiopathy. Acta Neuro-pathol. 117, 111–124

40. Bradshaw, E. M., Chibnik, L. B., Keenan, B. T., Ottoboni, L., Raj, T.,Tang, A., Rosenkrantz, L. L., Imboywa, S., Lee, M., Von Korff, A.,Morris, M. C., Evans, D. A., Johnson, K., Sperling, R. A., Schneider,J. A., Bennett, D. A., and De Jager, P. L.; Alzheimer DiseaseNeuroimaging Initiative. (2013) CD33 Alzheimer’s disease locus:altered monocyte function and amyloid biology. Nat. Neurosci. 16,848–850

41. Griciuc, A., Serrano-Pozo, A., Parrado, A. R., Lesinski, A. N., Asselin,C.N.,Mullin,K.,Hooli, B., Choi, S.H.,Hyman,B.T., andTanzi, R. E.(2013) Alzheimer’s disease risk gene CD33 inhibits microglialuptake of amyloid beta. Neuron 78, 631–643

42. McGeer, P. L., Schulzer, M., andMcGeer, E. G. (1996) Arthritis andanti-inflammatory agents as possible protective factors for Alz-heimer’s disease: a review of 17 epidemiologic studies. Neurology 47,425–432

43. McGeer, P. L., Kawamata, T., Walker, D. G., Akiyama, H., Tooyama,I., andMcGeer, E. G. (1993)Microglia in degenerative neurologicaldisease. Glia 7, 84–92

44. Hong, S., Beja-Glasser, V. F., Nfonoyim, B. M., Frouin, A., Li, S.,Ramakrishnan, S., Merry, K. M., Shi, Q., Rosenthal, A., Barres, B. A.,Lemere,C.A., Selkoe,D. J., andStevens,B. (2016)Complement andmicroglia mediate early synapse loss in Alzheimer mouse models.Science 352, 712–716

45. Saresella, M., Marventano, I., Calabrese, E., Piancone, F., Rainone,V., Gatti, A., Alberoni, M., Nemni, R., and Clerici, M. (2014) Acomplex proinflammatory role for peripheral monocytes inAlzheimer’s disease. J. Alzheimers Dis. 38, 403–413

46. Hjorth, E., Zhu, M., Toro, V. C., Vedin, I., Palmblad, J., Cederholm,T., Freund-Levi, Y., Faxen-Irving, G., Wahlund, L. O., Basun, H.,Eriksdotter, M., and Schultzberg, M. (2013) Omega-3 fatty acidsenhancephagocytosis ofAlzheimer’s disease-related amyloid-b42byhuman microglia and decrease inflammatory markers. J. AlzheimersDis. 35, 697–713

47. Mizwicki,M.T.,Menegaz,D.,Zhang, J.,Barrientos-Duran,A.,Tse, S.,Cashman, J. R., Griffin, P. R., and Fiala, M. (2012) Genomic andnongenomic signaling induced by 1a,25(OH)2-vitamin D3 pro-motes the recovery of amyloid-b phagocytosis byAlzheimer’s diseasemacrophages. J. Alzheimers Dis. 29, 51–62

48. Fiala, M., Halder, R. C., Sagong, B., Ross, O., Sayre, J., Porter, V., andBredesen, D. E. (2015) n-3 Supplementation increases amyloid-bphagocytosis and resolvin D1 in patients with minor cognitiveimpairment. FASEB J. 29, 2681–2689

49. Famenini, S., Rigali, E. A., Olivera-Perez, H. M., Dang, J., Chang,M. T., Halder, R., Rao, R. V., Pellegrini, M., Porter, V., Bredesen, D.,and Fiala, M. (2017) Increased intermediate M1-M2 macrophagepolarization and improved cognition in mild cognitive impairmentpatients on n-3 supplementation. FASEB J. 31, 148–160

50. Lam, L., Halder, R. C.,Montoya, D. J., Rubbi, L., Rinaldi, A., Sagong,B.,Weitzman, S., Rubattino,R., Singh,R.R., Pellegrini,M., andFiala,M. (2015) Anti-inflammatory therapies of amyotrophic lateral scle-rosis guided by immune pathways. Am. J. Neurodegener. Dis. 4, 28–39

51. Lam, L., Chin, L., Halder, R. C., Sagong, B., Famenini, S., Sayre, J.,Montoya, D., Rubbi, L., Pellegrini, M., and Fiala, M. (2016)Epigenetic changes in T-cell and monocyte signatures and

8 Vol. 31 August 2017 FIALA ET AL.The FASEB Journal x www.fasebj.org Vol.,  No. , pp:, April, 2017The FASEB Journal. 169.237.169.81 to IP www.fasebj.orgDownloaded from

Page 9: Modulation of innate immunity of patients with Alzheimer’s ... · THE JOURNAL † REVIEW † Modulation of innate immunity of patients with Alzheimer’s disease by omega-3 fatty

production of neurotoxic cytokines in ALS patients. FASEB J. 30,3461–3473

52. Serhan,C.N., Chiang,N., andDalli, J. (2015)The resolution codeofacute inflammation: novel pro-resolving lipid mediators in resolu-tion. Semin. Immunol. 27, 200–215

53. Serhan, C. N. (2010) Novel lipid mediators and resolutionmechanisms in acute inflammation: to resolve or not? Am. J.Pathol. 177, 1576–1591

54. Haeggstrom, J. Z., and Funk, C. D. (2011) Lipoxygenase andleukotriene pathways: biochemistry, biology, and roles in disease.Chem. Rev. 111, 5866–5898

55. Zhu,M.,Wang,X.,Hjorth, E., Colas, R.A., Schroeder, L.,Granholm,A. C., Serhan, C. N., and Schultzberg, M. (2016) Pro-resolving lipidmediators improve neuronal survival and increase Ab42 phagocy-tosis.Mol. Neurobiol. 53, 2733–2749

56. Fullerton, J. N., and Gilroy, D. W. (2016) Resolution ofinflammation: a new therapeutic frontier. Nat. Rev. Drug Discov. 15,551–567

57. Levy, B. D., Clish, C. B., Schmidt, B., Gronert, K., and Serhan,C. N. (2001) Lipid mediator class switching during acuteinflammation: signals in resolution. Nat. Immunol. 2, 612–619

58. Serhan, C. N., Dalli, J., Colas, R. A., Winkler, J. W., and Chiang, N.(2015) Protectins and maresins: new pro-resolving families of me-diators in acute inflammation and resolutionbioactivemetabolome.Biochim. Biophys. Acta 1851, 397–413

59. Serhan,C.N.,Dalli, J.,Karamnov, S.,Choi,A., Park,C.K.,Xu,Z.Z., Ji,R. R., Zhu, M., and Petasis, N. A. (2012) Macrophage proresolvingmediatormaresin1 stimulates tissue regenerationandcontrolspain.FASEB J. 26, 1755–1765

60. Dalli, J., Chiang, N., and Serhan, C. N. (2014) Identification of 14-series sulfido-conjugated mediators that promote resolution of in-fection and organ protection. Proc. Natl. Acad. Sci. USA 111,E4753–E4761

61. Dalli, J., Ramon, S., Norris, P. C., Colas, R. A., and Serhan, C. N.(2015) Novel proresolving and tissue-regenerative resolvin andprotectin sulfido-conjugated pathways. FASEB J. 29, 2120–2136

62. Krishnamoorthy, S., Recchiuti, A., Chiang, N., Yacoubian, S., Lee,C. H., Yang, R., Petasis, N. A., and Serhan, C. N. (2010) Resolvin D1binds human phagocytes with evidence for proresolving receptors.Proc. Natl. Acad. Sci. USA 107, 1660–1665

63. Terrando, N., Gomez-Galan, M., Yang, T., Carlstrom, M.,Gustavsson, D., Harding, R. E., Lindskog, M., and Eriksson, L. I.(2013) Aspirin-triggered resolvin D1 prevents surgery-induced cog-nitive decline. FASEB J. 27, 3564–3571

64. Chiang, N., Dalli, J., Colas, R. A., and Serhan, C. N. (2015)Identification of resolvin D2 receptor mediating resolution ofinfections and organ protection. J. Exp. Med. 212, 1203–1217

65. Arita, M., Bianchini, F., Aliberti, J., Sher, A., Chiang, N., Hong, S.,Yang, R., Petasis, N. A., and Serhan, C. N. (2005) Stereochemicalassignment, antiinflammatory properties, and receptor for theomega-3 lipid mediator resolvin E1. J. Exp. Med. 201, 713–722

66. Dalli, J., Zhu,M.,Vlasenko,N.A.,Deng,B.,Haeggstrom, J.Z., Petasis,N. A., and Serhan, C. N. (2013) The novel 13S,14S-epoxy-maresin isconverted by human macrophages to maresin 1 (MaR1), inhibitsleukotriene A4 hydrolase (LTA4H), and shifts macrophage phe-notype. FASEB J. 27, 2573–2583

67. Bazan, N. G. (2013) The docosanoid neuroprotectin D1 induceshomeostatic regulation of neuroinflammation and cell survival.Prostaglandins Leukot. Essent. Fatty Acids 88, 127–129

68. Spite, M., Norling, L. V., Summers, L., Yang, R., Cooper, D., Petasis,N.A., Flower,R. J., Perretti,M., andSerhan,C.N. (2009)ResolvinD2is a potent regulator of leukocytes and controls microbial sepsis.Nature 461, 1287–1291

69. Hasturk, H., Kantarci, A., Goguet-Surmenian, E., Blackwood,A., Andry, C., Serhan, C. N., and Van Dyke, T. E. (2007)Resolvin E1 regulates inflammation at the cellular and tissuelevel and restores tissue homeostasis in vivo. J. Immunol. 179,7021–7029

70. Rajasagi, N. K., Reddy, P. B., Mulik, S., Gjorstrup, P., and Rouse,B. T. (2013) Neuroprotectin D1 reduces the severity of herpessimplex virus-induced corneal immunopathology. Invest. Oph-thalmol. Vis. Sci. 54, 6269–6279

71. Borgeson, E., Johnson,A.M., Lee, Y. S., Till, A., Syed,G.H., Ali-Shah,S. T., Guiry, P. J., Dalli, J., Colas, R. A., Serhan, C. N., Sharma, K., andGodson, C. (2015) Lipoxin A4 attenuates obesity-induced adiposeinflammation and associated liver and kidney disease.Cell Metab. 22,125–137

72. Wu, Y., Ye, X.H., Guo, P. P., Xu, S. P., Wang, J., Yuan, S. Y., Yao, S. L.,and Shang, Y. (2010) Neuroprotective effect of lipoxin A4 methylester in a rat model of permanent focal cerebral ischemia. J. Mol.Neurosci. 42, 226–234

73. Wu, J., Wang, A., Min, Z., Xiong, Y., Yan, Q., Zhang, J., Xu, J., andZhang, S. (2011) Lipoxin A4 inhibits the production ofproinflammatory cytokines induced by b-amyloid in vitro and invivo. Biochem. Biophys. Res. Commun. 408, 382–387

74. Norling, L. V., Headland, S. E., Dalli, J., Arnardottir, H.H.,Haworth,O., Jones, H. R., Irimia, D., Serhan, C. N., and Perretti, M. (2016)Proresolving and cartilage-protective actions of resolvin D1 in in-flammatory arthritis. JCI Insight 1, e85922

75. Greene, E. R., Huang, S., Serhan, C. N., and Panigrahy, D. (2011)Regulation of inflammation in cancer by eicosanoids. ProstaglandinsOther Lipid Mediat. 96, 27–36

76. Elajami, T. K., Colas, R. A., Dalli, J., Chiang, N., Serhan, C. N., andWelty, F. K. (2016) Specialized proresolving lipid mediators inpatients with coronary artery disease and their potential for clotremodeling. FASEB J. 30, 2792–2801

77. Wagner, K., Inceoglu, B., and Hammock, B. D. (2011) Solubleepoxide hydrolase inhibition, epoxygenated fatty acids andnociception. Prostaglandins Other Lipid Mediat. 96, 76–83

78. Morisseau, C., Inceoglu, B., Schmelzer, K., Tsai, H. J., Jinks, S. L.,Hegedus, C. M., and Hammock, B. D. (2010) Naturally occurringmonoepoxides of eicosapentaenoic acid and docosahexaenoic acidare bioactive antihyperalgesic lipids. J. Lipid Res. 51, 3481–3490

79. Kodani, S. D., and Hammock, B. D. (2015) The 2014 Bernard B.Brodie award lecture-epoxide hydrolases: drug metabolism to ther-apeutics for chronic pain. Drug Metab. Dispos. 43, 788–802

80. Sudhahar, V., Shaw, S., and Imig, J. D. (2010) Epoxyeicosatrienoicacid analogs and vascular function. Curr. Med. Chem. 17, 1181–1190

81. Schmelzer, K. R., Inceoglu, B., Kubala, L., Kim, I. H., Jinks, S. L.,Eiserich, J. P., and Hammock, B. D. (2006) Enhancement ofantinociception by coadministration of nonsteroidal anti-inflammatory drugs and soluble epoxide hydrolase inhibitors. Proc.Natl. Acad. Sci. USA 103, 13646–13651

82. Schmelzer,K.R., Kubala, L.,Newman, J.W.,Kim, I.H., Eiserich, J. P.,and Hammock, B. D. (2005) Soluble epoxide hydrolase is atherapeutic target for acute inflammation. Proc. Natl. Acad. Sci.USA 102, 9772–9777

83. Zeilhofer, H. U. (2007) Prostanoids in nociception and pain.Biochem. Pharmacol. 73, 165–174

84. Serhan, C. N., Chiang, N., Dalli, J., and Levy, B. D. (2014) Lipidmediators in the resolution of inflammation. Cold Spring Harb.Perspect. Biol. 7, a016311

85. Wagner, K., Inceoglu, B., Gill, S. S., and Hammock, B. D. (2011)Epoxygenated fatty acids and soluble epoxide hydrolase inhibition:novelmediators of pain reduction. J. Agric. Food Chem. 59, 2816–2824

86. Inceoglu, B., Wagner, K., Schebb, N. H., Morisseau, C., Jinks, S. L.,Ulu, A., Hegedus, C., Rose, T., Brosnan, R., and Hammock, B. D.(2011) Analgesia mediated by soluble epoxide hydrolase inhibitorsis dependent on cAMP. Proc. Natl. Acad. Sci. USA 108, 5093–5097

87. Inceoglu, B., Bettaieb, A., Trindade da Silva, C. A., Lee, K. S., Haj,F. G., and Hammock, B. D. (2015) Endoplasmic reticulum stress inthe peripheral nervous system is a significant driver of neuropathicpain. Proc. Natl. Acad. Sci. USA 112, 9082–9087

88. Iliff, J. J., Jia, J., Nelson, J., Goyagi, T., Klaus, J., and Alkayed, N. J.(2010) Epoxyeicosanoid signaling in CNS function and disease.Prostaglandins Other Lipid Mediat. 91, 68–84

89. Iliff, J. J., Wang, R., Zeldin, D. C., and Alkayed, N. J. (2009)Epoxyeicosanoids as mediators of neurogenic vasodilation incerebral vessels. Am. J. Physiol. Heart Circ. Physiol. 296, H1352–H1363

90. Nelson, J. W., Young, J. M., Borkar, R. N., Woltjer, R. L., Quinn, J. F.,Silbert, L. C., Grafe, M. R., and Alkayed, N. J. (2014) Role of solubleepoxide hydrolase in age-related vascular cognitive decline. Prosta-glandins Other Lipid Mediat. 113-115, 30–37

91. Ren, Q., Ma, M., Ishima, T., Morisseau, C., Yang, J., Wagner, K. M.,Zhang, J. C., Yang, C., Yao, W., Dong, C., Han, M., Hammock, B. D.,and Hashimoto, K. (2016) Gene deficiency and pharmacologicalinhibition of soluble epoxide hydrolase confers resilience torepeated social defeat stress. Proc. Natl. Acad. Sci. U. S. A. 113,E1944–E1952

92. Ma,M.,Ren,Q., Fujita, Y., Ishima,T., Zhang, J.C., andHashimoto,K.(2013) Effects of AS2586114, a soluble epoxide hydrolase inhibitor,on hyperlocomotion and prepulse inhibition deficits in mice afteradministration of phencyclidine. Pharmacol. Biochem. Behav. 110,98–103

MODULATION OF IMMUNITY IN ALZHEIMER’S DISEASE 9 Vol.,  No. , pp:, April, 2017The FASEB Journal. 169.237.169.81 to IP www.fasebj.orgDownloaded from

Page 10: Modulation of innate immunity of patients with Alzheimer’s ... · THE JOURNAL † REVIEW † Modulation of innate immunity of patients with Alzheimer’s disease by omega-3 fatty

93. Bazan, N. G., Calandria, J. M., and Serhan, C. N. (2010) Rescueand repair during photoreceptor cell renewal mediated bydocosahexaenoic acid-derived neuroprotectin D1. J. Lipid Res. 51,2018–2031

94. Lukiw, W. J., Cui, J. G., Marcheselli, V. L., Bodker, M., Botkjaer, A.,Gotlinger, K., Serhan, C. N., and Bazan, N. G. (2005) A role fordocosahexaenoic acid-derived neuroprotectin D1 in neural cellsurvival and Alzheimer disease. J. Clin. Invest. 115, 2774–2783

95. Zhao, Y., Calon, F., Julien, C., Winkler, J. W., Petasis, N. A., Lukiw,W. J., and Bazan, N. G. (2011) Docosahexaenoic acid-derived neu-roprotectin D1 induces neuronal survival via secretase- and PPARg-mediated mechanisms in Alzheimer’s disease models. PLoS One 6,e15816

96. Medeiros,R., Kitazawa,M., Passos,G.F., Baglietto-Vargas,D.,Cheng,D., Cribbs, D.H., andLaFerla, F.M. (2013)Aspirin-triggered lipoxinA4 stimulates alternative activation of microglia and reduces Alz-heimer disease-like pathology in mice. Am. J. Pathol. 182, 1780–1789

97. Fiala, M., Chattopadhay, M., La Cava, A., Tse, E., Liu, G., Lourenco,E., Eskin, A., Liu, P. T., Magpantay, L., Tse, S., Mahanian, M.,Weitzman, R., Tong, J., Nguyen, C., Cho, T., Koo, P., Sayre, J.,Martinez-Maza, O., Rosenthal, M. J., and Wiedau-Pazos, M. (2010)IL-17A is increased in the serum and in spinal cord CD8 and mastcells of ALS patients. J. Neuroinflammation 7, 76–90

98. Avagyan, H., Goldenson, B., Tse, E., Masoumi, A., Porter, V.,Wiedau-Pazos, M., Sayre, J., Ong, R., Mahanian, M., Koo, P., Bae, S.,Micic, M., Liu, P. T., Rosenthal, M. J., and Fiala, M. (2009) Immuneblood biomarkers of Alzheimer disease patients. J. Neuroimmunol.210, 67–72

99. Sica, A., and Mantovani, A. (2012) Macrophage plasticity andpolarization: in vivo veritas. J. Clin. Invest. 122, 787–795

100. Martinez, F.O., andGordon, S. (2014) TheM1 andM2paradigmofmacrophage activation: time for reassessment. F1000Prime Rep. 6, 13

101. Xue, J., Schmidt, S.V., Sander, J.,Draffehn,A., Krebs,W.,Quester, I.,DeNardo, D., Gohel, T. D., Emde,M., Schmidleithner, L., Ganesan,H., Nino-Castro, A., Mallmann, M. R., Labzin, L., Theis, H., Kraut,M., Beyer, M., Latz, E., Freeman, T. C., Ulas, T., and Schultze, J. L.(2014) Transcriptome-based network analysis reveals a spectrummodel of human macrophage activation. Immunity 40, 274–288

102. Mantovani, A., Sozzani, S., Locati, M., Allavena, P., and Sica, A.(2002) Macrophage polarization: tumor-associatedmacrophages asa paradigm for polarized M2 mononuclear phagocytes. TrendsImmunol. 23, 549–555

103. Takeuchi, O., and Akira, S. (2010) Pattern recognition receptorsand inflammation. Cell 140, 805–820

104. Satoh, T., Takeuchi, O., Vandenbon, A., Yasuda, K., Tanaka, Y.,Kumagai, Y., Miyake, T., Matsushita, K., Okazaki, T., Saitoh, T.,Honma, K., Matsuyama, T., Yui, K., Tsujimura, T., Standley, D. M.,Nakanishi, K., Nakai, K., and Akira, S. (2010) The Jmjd3-Irf4 axisregulates M2 macrophage polarization and host responses againsthelminth infection. Nat. Immunol. 11, 936–944

105. Martinez-Nunez,R.T.,Louafi, F., andSanchez-Elsner,T. (2011)Theinterleukin13(IL-13)pathway inhumanmacrophages ismodulatedby microRNA-155 via direct targeting of interleukin 13 receptoralpha1 (IL13Ralpha1). J. Biol. Chem. 286, 1786–1794

106. Biswas, S. K., and Mantovani, A. (2010) Macrophage plasticity andinteraction with lymphocyte subsets: cancer as a paradigm. Nat.Immunol. 11, 889–896

107. Fiala, M., Cribbs, D. H., Rosenthal, M., and Bernard, G. (2007)Phagocytosis of amyloid-beta and inflammation: two faces of innateimmunity in Alzheimer’s disease. J. Alzheimers Dis. 11, 457–463

108. Fiala,M.,Mahanian,M., Rosenthal,M.,Mizwicki,M.T., Tse, E., Cho,T., Sayre, J., Weitzman, R., and Porter, V. (2011) MGAT3 mRNA: abiomarker for prognosis and therapy of Alzheimer’s disease byvitamin D and curcuminoids. J. Alzheimers Dis. 25, 135–144

109. Jha, A. K., Huang, S. C., Sergushichev, A., Lampropoulou, V.,Ivanova, Y., Loginicheva, E., Chmielewski, K., Stewart, K. M., Ashall,J., Everts, B., Pearce, E. J., Driggers, E. M., and Artyomov, M. N.(2015)Network integrationofparallelmetabolic and transcriptionaldata reveals metabolic modules that regulate macrophagepolarization. Immunity 42, 419–430

110. Kizuka, Y., Kitazume, S., Fujinawa,R., Saito, T., Iwata,N., Saido, T.C.,Nakano,M., Yamaguchi, Y., Hashimoto, Y., Staufenbiel, M., Hatsuta,H.,Murayama, S.,Manya,H., Endo,T., andTaniguchi,N. (2015)Anaberrant sugarmodification of BACE1 blocks its lysosomal targetingin Alzheimer’s disease. EMBO Mol. Med. 7, 175–189

111. Gonzalez-Periz, A., Horrillo, R., Ferre, N., Gronert, K., Dong, B.,Moran-Salvador, E., Titos, E., Martınez-Clemente, M., Lopez-Parra,

M., Arroyo, V., and Claria, J. (2009) Obesity-induced insulin re-sistance and hepatic steatosis are alleviated by omega-3 fatty acids: arole for resolvins and protectins. FASEB J. 23, 1946–1957

112. Hellmann, J., Tang, Y., Kosuri, M., Bhatnagar, A., and Spite, M.(2011) ResolvinD1 decreases adipose tissuemacrophage accumu-lation and improves insulin sensitivity in obese-diabeticmice. FASEBJ. 25, 2399–2407

113. Dalli, J., Winkler, J. W., Colas, R. A., Arnardottir, H., Cheng, C. Y.,Chiang, N., Petasis, N. A., and Serhan, C. N. (2013) Resolvin D3 andaspirin-triggered resolvin D3 are potent immunoresolvents. Chem.Biol. 20, 188–201

114. Vasconcelos, D. P., Costa, M., Amaral, I. F., Barbosa, M. A., Aguas,A. P., and Barbosa, J. N. (2015) Modulation of the inflammatoryresponse to chitosan through M2 macrophage polarization usingpro-resolution mediators. Biomaterials 37, 116–123

115. Dalli, J., and Serhan, C. N. (2012) Specific lipidmediator signaturesof human phagocytes: microparticles stimulate macrophageefferocytosis and pro-resolving mediators. Blood 120, e60–e72

116. Ramon, S., Dalli, J., Sanger, J. M., Winkler, J. W., Aursnes, M.,Tungen, J. E.,Hansen,T. V., and Serhan, C.N. (2016)TheProtectinPCTR1 is produced by human M2 macrophages and enhancesresolution of infectious inflammation. Am. J. Pathol. 186, 962–973

117. Masoodi, M., Kuda, O., Rossmeisl, M., Flachs, P., and Kopecky, J.(2015)Lipid signaling in adipose tissue: connecting inflammation&metabolism. Biochim. Biophys. Acta 1851, 503–518

118. Gottlicher,M.,Widmark, E., Li,Q., andGustafsson, J.A. (1992)Fattyacids activate a chimera of the clofibric acid-activated receptor andthe glucocorticoid receptor. Proc. Natl. Acad. Sci. USA 89, 4653–4657

119. Jump, D. B. (2008) N-3 polyunsaturated fatty acid regulation of he-patic gene transcription. Curr. Opin. Lipidol. 19, 242–247

120. Bordoni, A., Di Nunzio, M., Danesi, F., and Biagi, P. L. (2006)Polyunsaturated fatty acids: from diet to binding to ppars and othernuclear receptors. Genes Nutr. 1, 95–106

121. Chang, H. Y., Lee, H. N., Kim, W., and Surh, Y. J. (2015)Docosahexaenoic acid induces M2 macrophage polarization throughperoxisomeproliferator-activated receptor g activation.Life Sci. 120,39–47

122. Heras-Sandoval, D., Pedraza-Chaverri, J., and Perez-Rojas, J. M.(2016) Role of docosahexaenoic acid in themodulation of glial cellsin Alzheimer’s disease. J. Neuroinflammation 13, 61

123. Sperling, R., Salloway, S., Brooks, D. J., Tampieri, D., Barakos, J., Fox,N. C., Raskind, M., Sabbagh, M., Honig, L. S., Porsteinsson, A. P.,Lieberburg, I., Arrighi, H. M., Morris, K. A., Lu, Y., Liu, E., Gregg,K. M., Brashear, H. R., Kinney, G. G., Black, R., and Grundman, M.(2012) Amyloid-related imaging abnormalities in patients with Alz-heimer’s disease treated with bapineuzumab: a retrospective analy-sis. Lancet Neurol. 11, 241–249

124. Busche, M. A., Grienberger, C., Keskin, A. D., Song, B., Neumann,U., Staufenbiel, M., Forstl, H., and Konnerth, A. (2015) Decreasedamyloid-b and increased neuronal hyperactivity by immunotherapyin Alzheimer’s models. Nat. Neurosci. 18, 1725–1727

125. McGeer, P. L., andMcGeer, E.G. (2015)Targetingmicroglia for thetreatment of Alzheimer’s disease. Expert Opin. Ther. Targets 19,497–506

126. Bohrmann, B., Baumann, K., Benz, J., Gerber, F., Huber, W.,Knoflach, F., Messer, J., Oroszlan, K., Rauchenberger, R., Richter,W. F., Rothe, C., Urban, M., Bardroff, M., Winter, M., Nordstedt, C.,and Loetscher, H. (2012) Gantenerumab: a novel human anti-Abantibody demonstrates sustained cerebral amyloid-b binding andelicits cell-mediated removal of human amyloid-b. J. Alzheimers Dis.28, 49–69

127. Sevigny, J.,Chiao,P.,Bussiere,T.,Weinreb,P.H.,Williams,L.,Maier,M., Dunstan, R., Salloway, S., Chen, T., Ling, Y., O’Gorman, J., Qian,F., Arastu,M., Li,M.,Chollate, S., Brennan,M. S.,Quintero-Monzon,O., Scannevin, R.H., Arnold,H.M., Engber, T., Rhodes, K., Ferrero,J., Hang, Y., Mikulskis, A., Grimm, J., Hock, C., Nitsch, R. M., andSandrock, A. (2016)The antibody aducanumab reducesAb plaquesin Alzheimer’s disease. Nature 537, 50–56

128. Goel, A., and Aggarwal, B. B. (2010) Curcumin, the golden spicefrom Indian saffron, is a chemosensitizer and radiosensitizer fortumors and chemoprotector and radioprotector for normal organs.Nutr. Cancer 62, 919–930

129. Sarkar, S., Hewison, M., Studzinski, G. P., Li, Y. C., and Kalia, V.(2016) Role of vitamin D in cytotoxic T lymphocyte immunity topathogens and cancer. Crit. Rev. Clin. Lab. Sci. 53, 132–145

130. Martin, C. (2014) Diet and Nutrition in Dementia and Cognitive Decline,Elsevier, Boston, MA

10 Vol. 31 August 2017 FIALA ET AL.The FASEB Journal x www.fasebj.org Vol.,  No. , pp:, April, 2017The FASEB Journal. 169.237.169.81 to IP www.fasebj.orgDownloaded from

Page 11: Modulation of innate immunity of patients with Alzheimer’s ... · THE JOURNAL † REVIEW † Modulation of innate immunity of patients with Alzheimer’s disease by omega-3 fatty

131. Fiala,M., Terrando,N., andDalli, J. (2015) Specializedpro-resolvingmediators fromomega-3 fatty acids improve amyloid-b phagocytosisand regulate inflammation in patients with minor cognitive im-pairment. J. Alzheimers Dis. 48, 293–301

132. Annweiler, C., Dursun, E., Feron, F., Gezen-Ak, D., Kalueff, A. V.,Littlejohns, T., Llewellyn, D. J., Millet, P., Scott, T., Tucker, K. L.,Yilmazer, S., and Beauchet, O. (2015) ‘Vitamin D and cognition inolder adults’: updated international recommendations. J. Intern.Med. 277, 45–57

133. Jerneren, F., Elshorbagy, A. K., Oulhaj, A., Smith, S. M., Refsum, H.,and Smith, A. D. (2015) Brain atrophy in cognitively impairedelderly: the importance of long-chain n-3 fatty acids and B vitaminstatus ina randomizedcontrolled trial.Am. J. Clin.Nutr.102, 215–221

134. Goel, A., Kunnumakkara, A. B., and Aggarwal, B. B. (2008)Curcumin as “curecumin”: from kitchen to clinic. Biochem.Pharmacol. 75, 787–809

135. Masoumi, A., Goldenson, B., Ghirmai, S., Avagyan, H., Zaghi, J.,Abel, K., Zheng, X., Espinosa-Jeffrey, A., Mahanian, M., Liu, P. T.,Hewison, M., Mizwickie, M., Cashman, J., and Fiala, M. (2009)1alpha,25-dihydroxyvitamin D3 interacts with curcuminoids tostimulate amyloid-beta clearance by macrophages of Alzheimer’sdisease patients. J. Alzheimers Dis. 17, 703–717

136. Goozee, K. G., Shah, T. M., Sohrabi, H. R., Rainey-Smith, S. R.,Brown, B., Verdile, G., and Martins, R. N. (2016) Examining thepotential clinical value of curcumin in the prevention and diagnosisof Alzheimer’s disease. Br. J. Nutr. 115, 449–465

137. Cramer, P. E., Cirrito, J. R., Wesson, D. W., Lee, C. Y., Karlo, J. C.,Zinn, A. E., Casali, B. T., Restivo, J. L., Goebel, W. D., James, M. J.,Brunden, K. R., Wilson, D. A., and Landreth, G. E. (2012) ApoE-directed therapeutics rapidly clear b-amyloid and reverse deficits inAD mouse models. Science 335, 1503–1506

138. Landreth, G. E., Cramer, P. E., Lakner, M. M., Cirrito, J. R., Wesson,D. W., Brunden, K. R., and Wilson, D. A. (2013) Response tocomments on “ApoE-directed therapeutics rapidly clear beta-amyloid and reverse deficits in AD mouse models”. Science 340, 924

139. O’Hare, E., Jeggo, R., Kim, E.M., Barbour, B.,Walczak, J. S., Palmer,P., Lyons, T., Page, D., Hanna, D., Meara, J. R., Spanswick, D., Guo,J. P., McGeer, E. G., McGeer, P. L., and Hobson, P. (2016) Lack ofsupport for bexarotene as a treatment for Alzheimer’s disease.Neuropharmacology 100, 124–130

140. Turner, R. S., Thomas, R. G., Craft, S., van Dyck, C. H., Mintzer, J.,Reynolds, B. A., Brewer, J. B., Rissman, R. A., Raman, R., and Aisen,P. S.; Alzheimer’s Disease Cooperative Study. (2015) A randomized,double-blind, placebo-controlled trial of resveratrol for Alzheimerdisease. Neurology 85, 1383–1391

141. Pasinetti, G. M., Wang, J., Ho, L., Zhao, W., and Dubner, L. (2015)Roles of resveratrol and other grape-derived polyphenols in Alz-heimer’s disease prevention and treatment. Biochim. Biophys. Acta1852, 1202–1208

142. Yakeu, G., Butcher, L., Isa, S., Webb, R., Roberts, A. W., Thomas,A. W., Backx, K., James, P. E., and Morris, K. (2010) Low-intensityexercise enhances expression of markers of alternative activation incirculating leukocytes: roles of PPARg and Th2 cytokines. Athero-sclerosis 212, 668–673

143. Jicha, G. A., and Markesbery, W. R. (2010) Omega-3 fatty acids:potential role in themanagement of early Alzheimer’s disease. Clin.Interv. Aging 5, 45–61

144. Freund-Levi, Y., Eriksdotter-Jonhagen, M., Cederholm, T., Basun,H., Faxen-Irving, G., Garlind, A., Vedin, I., Vessby, B., Wahlund,L. O., and Palmblad, J. (2006) Omega-3 fatty acid treatment in 174patientswithmild tomoderate Alzheimer disease:OmegAD study: arandomized double-blind trial. Arch. Neurol. 63, 1402–1408

145. Kulzow, N., Witte, A. V., Kerti, L., Grittner, U., Schuchardt, J. P.,Hahn, A., and Floel, A. (2016) Impact of omega-3 fatty acid sup-plementation on memory functions in healthy older adults.J. Alzheimers Dis. 51, 713–725

146. Quinn, J. F., Raman, R., Thomas, R. G., Yurko-Mauro, K., Nelson,E.B.,VanDyck,C.,Galvin, J.E., Emond, J., Jack,C.R., Jr.,Weiner,M.,Shinto, L., and Aisen, P. S. (2010) Docosahexaenoic acidsupplementation and cognitive decline in Alzheimer disease: arandomized trial. JAMA 304, 1903–1911

147. Shinto, L., Quinn, J., Montine, T., Dodge, H. H., Woodward, W.,Baldauf-Wagner, S., Waichunas, D., Bumgarner, L., Bourdette, D.,Silbert, L., and Kaye, J. (2014) A randomized placebo-controlledpilot trial of omega-3 fatty acids and alpha lipoic acid in Alzheimer’sdisease. J. Alzheimers Dis. 38, 111–120

148. Burckhardt, M., Herke, M., Wustmann, T., Watzke, S., Langer, G.,and Fink, A. (2016) Omega-3 fatty acids for the treatment of de-mentia. Cochrane Database Syst. Rev. 4, CD009002

149. Morris, M. C., Brockman, J., Schneider, J. A., Wang, Y., Bennett,D. A., Tangney, C. C., and van de Rest, O. (2016) Association ofseafood consumption, brainmercury level, andAPOE e4 status withbrain neuropathology in older adults. JAMA 315, 489–497

150. Bredesen, D. E., Amos, E. C., Canick, J., Ackerley, M., Raji, C., Fiala,M., and Ahdidan, J. (2016) Reversal of cognitive decline inAlzheimer’s disease. Aging (Albany NY) 8, 1250–1258

151. Jackowski, S. A., Alvi, A. Z., Mirajkar, A., Imani, Z., Gamalevych, Y.,Shaikh, N. A., and Jackowski, G. (2015) Oxidation levels of NorthAmerican over-the-counter n-3 (omega-3) supplements and the in-fluence of supplement formulation and delivery form on evaluatingoxidative safety. J. Nutr. Sci. 4, e30

152. Lohcharoenkal, W., Wang, L., Chen, Y. C., and Rojanasakul, Y.(2014) Protein nanoparticles as drug delivery carriers for cancertherapy. BioMed Res. Int. 2014, 180549

153. Theendakara, V., Patent, A., Peters Libeu, C. A., Philpot, B., Flores,S.,Descamps,O., Poksay,K. S., Zhang,Q.,Cailing,G.,Hart,M., John,V., Rao, R. V., and Bredesen, D. E. (2013) Neuroprotective Sirtuinratio reversed by ApoE4. Proc. Natl. Acad. Sci. USA 110, 18303–18308

154. Annweiler, C., Llewellyn, D. J., and Beauchet, O. (2013) Low serumvitaminD concentrations in Alzheimer’s disease: a systematic reviewand meta-analysis. J. Alzheimers Dis. 33, 659–674

155. Oulhaj, A., Jerneren, F., Refsum,H., Smith, A. D., and de Jager, C. A.(2016) Omega-3 fatty acid status enhances the prevention of cog-nitive decline by B vitamins in mild cognitive impairment.J. Alzheimers Dis. 50, 547–557

156. Barnard, N. D., Bush, A. I., Ceccarelli, A., Cooper, J., de Jager, C. A.,Erickson, K. I., Fraser, G., Kesler, S., Levin, S. M., Lucey, B., Morris,M. C., and Squitti, R. (2014) Dietary and lifestyle guidelines for theprevention of Alzheimer’s disease. Neurobiol. Aging 35, S74–S78

157. Doody,R. S., Thomas,R.G., Farlow,M., Iwatsubo,T., Vellas, B., Joffe,S., Kieburtz, K., Raman, R., Sun, X., Aisen, P. S., Siemers, E.,Liu-Seifert,H., andMohs,R.; Alzheimer’sDiseaseCooperativeStudySteering Committee; Solanezumab Study Group. (2014) Phase 3trials of solanezumab for mild-to-moderate Alzheimer’s disease. N.Engl. J. Med. 370, 311–321

158. Salloway, S., Sperling,R., Fox,N.C.,Blennow,K.,Klunk,W.,Raskind,M., Sabbagh,M.,Honig,L. S., Porsteinsson, A. P., Ferris, S., Reichert,M., Ketter, N., Nejadnik, B., Guenzler, V., Miloslavsky, M., Wang, D.,Lu, Y., Lull, J., Tudor, I.C.,Liu,E.,Grundman,M., Yuen,E.,Black,R.,and Brashear, H. R.; Bapineuzumab 301 and 302 Clinical TrialInvestigators. (2014)Two phase 3 trials of bapineuzumab inmild-to-moderate Alzheimer’s disease. N. Engl. J. Med. 370, 322–333

159. Chew, E. Y., Clemons, T. E., Agron, E., Launer, L. J., Grodstein, F.,and Bernstein, P. S.; Age-Related Eye Disease Study 2 (AREDS2)Research Group. (2015) Effect of omega-3 fatty acids, lutein/zeaxanthin, or other nutrient supplementation on cognitive func-tion: the AREDS2 randomized clinical trial. JAMA 314, 791–801

160. Serhan, C. N. (1991) Lipoxins: eicosanoids carrying intra- andintercellular messages. J. Bioenerg. Biomembr. 23, 105–122

Received for publication January 27, 2017.Accepted for publication April 5, 2017.

MODULATION OF IMMUNITY IN ALZHEIMER’S DISEASE 11 Vol.,  No. , pp:, April, 2017The FASEB Journal. 169.237.169.81 to IP www.fasebj.orgDownloaded from

Page 12: Modulation of innate immunity of patients with Alzheimer’s ... · THE JOURNAL † REVIEW † Modulation of innate immunity of patients with Alzheimer’s disease by omega-3 fatty

10.1096/fj.201700065RAccess the most recent version at doi: published online April 18, 2017FASEB J 

  Milan Fiala, Gijs Kooij, Karen Wagner, et al.   disease by omega-3 fatty acidsModulation of innate immunity of patients with Alzheimer's

Subscriptions

  http://www.faseb.org/The-FASEB-Journal/Librarian-s-Resources.aspx

is online at The FASEB JournalInformation about subscribing to

Permissions

  http://www.fasebj.org/site/misc/copyright.xhtmlSubmit copyright permission requests at:

Email Alerts

  http://www.fasebj.org/cgi/alertsReceive free email alerts when new an article cites this article - sign up at

© FASEB

 Vol.,  No. , pp:, April, 2017The FASEB Journal. 169.237.169.81 to IP www.fasebj.orgDownloaded from