minimotifs dysfunction is pervasive in neurodegenerative

19
Featured Article Minimotifs dysfunction is pervasive in neurodegenerative disorders Q25 Surbhi Sharma a,b , Richard J. Young a,b , Jingchun Chen a , Xiangning Chen a,c , Edwin C. Oh a,d, **, Martin R. Schiller a,b,d, * a Nevada Institute of Personalized Medicine, Las Vegas, NV, USA b School of Life Sciences, Las Vegas, NV, USA c Departmentof Psychology, Las Vegas, NV, USA d School of Medicine, Las Vegas, NV, USA Abstract Q2 Minimotifs are modular contiguous peptide sequences in proteins that are important for posttrans- lational modifications, binding to other molecules, and trafficking to specific subcellular compart- ments. Some molecular functions of proteins in cellular pathways can be predicted from minimotif consensus sequences identified through experimentation. While a role for minimotifs in regulating signal transduction and gene regulation during disease pathogenesis (such as infectious diseases and cancer) is established, the therapeutic use of minimotif mimetic drugs is limited. In this review, we discuss a general theme identifying a pervasive role of minimotifs in the pathome- chanism of neurodegenerative diseases. Beyond their longstanding history in the genetics of familial neurodegeneration, minimotifs are also major players in neurotoxic protein aggregation, aberrant protein trafficking, and epigenetic regulation. Generalizing the importance of minimotifs in neurode- generative diseases offers a new perspective for the future study of neurodegenerative mechanisms and the investigation of new therapeutics. Ó 2018 The Authors. Published by Elsevier Inc. on behalf of the Alzheimer’s Association. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/ 4.0/). Keywords: Minimotif; Posttranslational modification; Trafficking; Binding; Aggregate; Epigenetics; Genetics; Histone code; GWAS 1. Introduction Q3 Minimotifs, also called short linear motifs, are short contiguous peptide sequences or sequence patterns that encode molecular functions. These functions range from the binding of a protein to other proteins and molecules, posttranslational modification (PTM) of a protein, or traf- ficking of a protein to a subcellular compartment. Two main minimotif databases, Minimotif Miner and the Eukary- otic Linear Motif resource, now house more than one million minimotif instances [1–6]. Recent proteome-wide analysis of minimotifs with 1000 genomes data determined that the vast majority of minimotifs are fixed in humans, suggesting their importance in cellular function [7,8]. At the fundamental level, each minimotif is defined as a sequence or sequence pattern in a source protein and an activity that connects the motif to a target protein. The target protein can be an enzyme for PTM, an interaction with another molecule such as a protein, or a trafficking receptor. The conservation of most minimotifs and their role in evolution suggests that they might render a significant vulnerability to diseases [5,7–9]. In the first comprehensive review of minimotifs in human diseases in 2007, our group noticed that minimotifs were involved in disease, particularly infectious diseases, and others have expanded on this observation [5,9,10]. There are at least 35 minimotifs mutated in more than 20 diseases, including both rare and common disorders [11]. These include the three Conflict of interest: The authors declare that they have no competing in- terests. *Corresponding author. Tel.: 702-895-5546; Fax: 702-895-5728. **Corresponding author. Tel.: 702-895-0509; Fax: --- Q1 . E-mail address: [email protected] (E.C.O.), martin.schiller@unlv. edu (M.R.S.) https://doi.org/10.1016/j.trci.2018.06.005 2352-8737/ Ó 2018 The Authors. Published by Elsevier Inc. on behalf of the Alzheimer’s Association. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/). FLA 5.5.0 DTD ĸ TRCI168_proof ĸ 25 July 2018 ĸ 9:16 am ĸ ce Alzheimer’s & Dementia: Translational Research & Clinical Interventions - (2018) 1-19 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109

Upload: others

Post on 21-Apr-2022

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Minimotifs dysfunction is pervasive in neurodegenerative

Q25

Q2

Q3

Q1

Alzheimer’s & Dementia: Translational Research & Clinical Interventions - (2018) 1-19

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

21

22

23

24

25

26

27

28

29

30

31

32

33

34

35

36

37

38

39

40

41

42

43

44

45

46

47

48

49

50

51

52

53

54

55

56

57

58

59

60

Featured Article

Minimotifs dysfunction is pervasive in neurodegenerative disorders

61

62

63

64

65

66

67

68

69

Surbhi Sharmaa,b, Richard J. Younga,b, Jingchun Chena, Xiangning Chena,c, Edwin C. Oha,d,**,Martin R. Schillera,b,d,*

aNevada Institute of Personalized Medicine, Las Vegas, NV, USAbSchool of Life Sciences, Las Vegas, NV, USA

cDepartment of Psychology, Las Vegas, NV, USAdSchool of Medicine, Las Vegas, NV, USA

70

71

72

Abstract Minimotifs are modular contiguous peptide sequences in proteins that are important for posttrans-

Conflict of interest

terests.

*Corresponding au

**Corresponding

E-mail address: e

edu (M.R.S.)

https://doi.org/10.1016

2352-8737/� 2018 T

license (http://creative

73

74

75

76

77

78

79

80

81

82

83

84

85

86

87

88

lational modifications, binding to other molecules, and trafficking to specific subcellular compart-ments. Some molecular functions of proteins in cellular pathways can be predicted fromminimotif consensus sequences identified through experimentation. While a role for minimotifs inregulating signal transduction and gene regulation during disease pathogenesis (such as infectiousdiseases and cancer) is established, the therapeutic use of minimotif mimetic drugs is limited. Inthis review, we discuss a general theme identifying a pervasive role of minimotifs in the pathome-chanism of neurodegenerative diseases. Beyond their longstanding history in the genetics of familialneurodegeneration, minimotifs are also major players in neurotoxic protein aggregation, aberrantprotein trafficking, and epigenetic regulation. Generalizing the importance of minimotifs in neurode-generative diseases offers a new perspective for the future study of neurodegenerative mechanismsand the investigation of new therapeutics.� 2018 The Authors. Published by Elsevier Inc. on behalf of the Alzheimer’s Association. This is anopen access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

89

90

Keywords: Minimotif; Posttranslational modification; Trafficking; Binding; Aggregate; Epigenetics; Genetics; Histone code;

91

GWAS

92

93

94

95

96

97

98

99

100

101

102

103

104

105

106

1. Introduction

Minimotifs, also called short linear motifs, are shortcontiguous peptide sequences or sequence patterns thatencode molecular functions. These functions range fromthe binding of a protein to other proteins and molecules,posttranslational modification (PTM) of a protein, or traf-ficking of a protein to a subcellular compartment. Twomain minimotif databases, Minimotif Miner and the Eukary-otic LinearMotif resource, now house more than one millionminimotif instances [1–6]. Recent proteome-wide analysis

: The authors declare that they have no competing in-

thor. Tel.: 702-895-5546; Fax: 702-895-5728.

author. Tel.: 702-895-0509; Fax: ---.

[email protected] (E.C.O.), martin.schiller@unlv.

/j.trci.2018.06.005

he Authors. Published by Elsevier Inc. on behalf of the Alzhe

commons.org/licenses/by-nc-nd/4.0/).

FLA 5.5.0 DTD � TRCI168_proof

107

108

109

of minimotifs with 1000 genomes data determined that thevast majority of minimotifs are fixed in humans, suggestingtheir importance in cellular function [7,8]. At thefundamental level, each minimotif is defined as a sequenceor sequence pattern in a source protein and an activity thatconnects the motif to a target protein. The target proteincan be an enzyme for PTM, an interaction with anothermolecule such as a protein, or a trafficking receptor.

The conservation of most minimotifs and their role inevolution suggests that they might render a significantvulnerability to diseases [5,7–9]. In the first comprehensivereview of minimotifs in human diseases in 2007, our groupnoticed that minimotifs were involved in disease,particularly infectious diseases, and others have expandedon this observation [5,9,10]. There are at least 35minimotifs mutated in more than 20 diseases, includingboth rare and common disorders [11]. These include the three

imer’s Association. This is an open access article under the CC BY-NC-ND

� 25 July 2018 � 9:16 am � ce

Page 2: Minimotifs dysfunction is pervasive in neurodegenerative

Q4

Q5

S. Sharma et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions - (2018) 1-192

110

111

112

113

114

115

116

117

118

119

120

121

122

123

124

125

126

127

128

129

130

131

132

133

134

135

136

137

138

139

140

141

142

143

144

145

146

147

148

149

150

151

152

153

154

155

156

157

158

159

160

161

162

163

164

165

166

167

168

169

170

171

172

173

174

175

176

177

178

179

180

181

182

183

184

185

186

187

188

189

190

191

192

193

194

195

196

197

198

199

200

201

202

203

204

205

206

207

208

209

210

211

212

213

214

215

216

217

218

219

220

221

222

223

224

225

226

227

228

229

230

231

232

233

234

235

236

237

general classes; binding, PTMs, and trafficking minimotifs.Several annotations for diseases are listed and described onthe Eukaryotic Linear Motif resources website. Approxi-mately 0.1% of missense mutations in the COSMIC somaticcancer mutation database overlap with a minimotif. There are100s of minimotifs encoded by viruses that are used to hijackhost cell processes, several essential for the viral life cycle.

Another line of evidence for the importance of minimotifsin disease is the emergence of Food and Drug Administration(FDA)-approved minimotif mimetic drug therapeutics [11].For example, protein kinase inhibitor drugs such as Gleevec(imatinib mesylate), Iressa (gefitinib), Sprycel (dasatinib),and Stutnet (sunitinib) among others block phosphorylationof minimotifs and have become useful for cancer treatmentand immunosuppression [12]. Drugs that block different pro-teases that cleave minimotifs such as Lotensin (benazepril),Novastan (argatroban), Januvia (sitagliptin), and many HIVprotease inhibitors are also minimotif-directed therapeutics.Peptide hormones tend to have core clusters of amino acidsthat bind to receptors. For example, peptide therapeutics orchemical agonists such as Supprelin LA (histrelin) for theGonadotropin Releasing Hormone receptor, Byetta (exena-tide) for the glucagon-like peptide 1 receptor, SomatulineDepot (lanreotide) for the somatostatin receptors, and opiatessuch as Demerol (meperidine) for opioid receptors mimic thedeterminants of this core cluster in interaction with receptors.There are also antiviral drugs to several lipidation enzymesthat modify minimotifs, and tirofiban is a drug that mimicsthe Arg-Gly-Asp ligand for integrins.

In this review, by exploring the specific role of minimotifsin NDs, we consolidate significant evidence that supports ourhypothesis that minimotifs have distinct and generalizablefunctional roles in ND etiology. Minimotifs are the key con-nections in the cellular molecular network, so it is not surpris-ing that they are vulnerable to pathological dysfunction andthat all NDs have multiple dysfunctions of minimotifs. Byquestioning whether the modification of minimotifs is causalor a consequence of other precipitating events, we recognizeboth are contributing factors to NDs. From the causal perspec-tive, some of the familial genes in NDs have mutations thatdisrupt minimotifs or minimotif targets. Minimotifs mayalso be causal through PTM of the histone code and epige-netics. Other, less direct roles of minimotifs in pathogenesisare through protein trafficking, PTMs, neurotoxic protein ag-gregation, and aggregate clearance.

Because there are approximately 100 NDs known [13], inthis review, we have focused on only the major NDs: Alz-heimer’s disease (AD), Parkinson’s disease (PD), Hunting-ton’s disease (HD), amyotrophic lateral sclerosis (ALS),frontotemporal dementia (FTD), and tauopathies, althoughwhere relevant, other NDs are mentioned.

6

238

239

240

241

242

243

2. Types of minimotifs in neurodegenerative diseases

All three types of minimotifs: binding, modifying, andtrafficking, have important roles in neurotoxic aggregate

FLA 5.5.0 DTD � TRCI168_proof

formation and clearance, epigenetics through the histonecode, protein trafficking, and PTMs of ND-related proteins.Some of the genes with familial inheritance in NDs harbormutation in minimotifs or enzymes that modify minimotifs,as summarized in Tables 1 and 2 and discussed below.

2.1. PTM minimotifs in neurodegenerative diseases

Previous reviews of NDs summarize the important rolesof PTMs, one general category of minimotifs. There aremore than 500 types of covalent modifications, with most,if not all proteins in the human proteome having one ormore modifications [14]. These PTMs perturb local andglobal structural changes thereby altering protein binding,trafficking, half-lives, activities, and signaling. Therefore,it is not surprising that several of these modifications havesubstantial roles in the pathology of NDs.

2.1.1. Glycosylation/glycationHalf of all proteins inmost cell types undergoglycosylation

and areN-glycosylated at theminimal consensus sequenceNx[S/T] and/or O-glycosylated on Ser or Thr with no specificsequence determinants [15–17]. Protein glycosylationstabilizes protein structural folds and facilitates proteintrafficking, protein quality control, receptor activation, andendocytosis [17,18]. Human mutations in N-glycosylationsites can result in severe disease pathology; for example, afamilial T183A mutation in a NxT glycosylation minimotifin a prion protein (PrP) can result in Creutzfeldt-Jakob disease(CJD) [11,19–22].

Curiously, there are also several germline mutationsimmediately juxtaposed to a N-glycosylation consensussequence: a P504L mutation in WFS1 of Wolframs syn-drome, E196K, V180I in PRP for CJD (P04156), andE196K also in Gerstmann-Straussler disease [23–25].However, whether these mutations alter glycosylation andinfluence pathogenesis is not yet clear.

Also unclear is the observation of altered glycosylation inNDs, a likely downstream event or epiphenomena. On aglobal scale, proteomic analyses of AD brains identified131 GlcNacylation sites in 81 proteins that revealed alteredglycosylation. Another global study on the brain and seraisolated from the HD transgenic mice identified differencesin the amount and the pattern of glycans in mice showing pa-thology [26]. Aberrant glycosylation of an additional tenglycosylated proteins in NDs (AD, PD, and HD) was sum-marized previously [27]. Acetylcholinesterase is abnormallyglycosylated in both CJD and AD [27,28]. Nonenzymaticglycation and aberrant glycosylation of tau are prevalent inAD and FTD [29–32].

2.1.2. PhosphorylationProtein phosphorylation of Ser Q, Thr, or Tyr residues is

biologically significant throughout development of disease.During the pathogenesis of NDs, aberrant phosphorylationresults in the misfolding and aggregation of neurotoxic

� 25 July 2018 � 9:16 am � ce

Page 3: Minimotifs dysfunction is pervasive in neurodegenerative

Table 1

Neurodegenerative disorder minimotif summary

NDs

Minimotifs

Aggregate/gene Familial genes Activities References

Alzheimer’s

disease (AD)

b-amyloid plaques,

neurofibrillary

tangles/APP,

MAPT

APP, PSEN1,

PSEN2, MAPT

Proteolysis

Sumoylation

Glycosylation

Lipidation

Acetylation

Methylation

Oxidative PTM

Ubiquitylation

Citrullination

Trafficking

Binding

[24,30,89,98,108,

153,156,164,

168,175,178,

269–273]

Amyotrophic

lateral sclerosis

(ALS)

Hyaline

inclusions/SOD1

SOD1 Ubiquitylation

Sumoylation

Phosphorylation

Lipidation

Oxidation

S-nitrosylation

S-glutathionylation

Trafficking

[82,85–87,

108,274–277]

Batten (neuronal

ceroid

lipofuscinosis)

Aggregation/CSPa Lipidation [63]

Creutzfeldt-Jakob Prion plaques/(PrP) PRNP Glycosylation

Trafficking

Phosphorylation

Acetylation

Methylation

Ubiquitylation

Citrullination

Binding

[10,23,99,

175,262,

278–281]

Frontotemporal

dementia/

degeneration (FTD)

Neurofibrillary

tangles/tau

tau Phosphorylation

Sumoylation

Ubiquitylation

Glycosylation

[27,32,

33,74,120]

Huntington’s

disease (HD)

Neuronal

inclusions/HTT

HTT Sumoylation

Glycosylation

Ubiquitylation

Proteolysis

Trafficking

[52,101,

108,273,282]

Parkinson’s

Disease (PD)

Lewy bodies/SNCA SNCA

LRRK2

GBA

VPS35

EIF4G

DNAJC13

PRKN

Proteolysis

Sumoylation

Glycosylation

Lipidation

Acetylation

Methylation

Oxidative PTM

Ubiquitylation

Phosphorylation

Citrullination,

Trafficking

[34,88,96,

108,270,

272,273,

283–290]

Spinal and

bulbar

muscular

atrophy

(SBMA)

Aggregation/AR Sumoylation

Ubiquitylation

Phosphorylation

of non-aggregating tau

[108,291–294]

Spinocerebellar

ataxia (SA)

Neuronal

inclusions/SCA

SCA Sumoylation

Phosphorylation

Ubiquitylation

Trafficking

Proteolysis

[48,100,101,

108,174,

295,296]

(Continued )

FLA 5.5.0 DTD � TRCI168_proof � 25 July 2018 � 9:16 am � ce

S. Sharma et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions - (2018) 1-19 3

244

245

246

247

248

249

250

251

252

253

254

255

256

257

258

259

260

261

262

263

264

265

266

267

268

269

270

271

272

273

274

275

276

277

278

279

280

281

282

283

284

285

286

287

288

289

290

291

292

293

294

295

296

297

298

299

300

301

302

303

304

305

306

307

308

309

310

311

312

313

314

315

316

317

318

319

320

321

322

323

324

325

326

327

328

329

330

331

332

333

334

335

336

337

338

339

340

341

342

343

344

345

346

347

348

349

350

351

352

353

354

355

356

357

358

359

360

361

362

363

364

365

366

367

368

369

370

371

372

373

374

375

376

377

Page 4: Minimotifs dysfunction is pervasive in neurodegenerative

Table 1

Neurodegenerative disorder minimotif summary (Continued )

NDs

Minimotifs

Aggregate/gene Familial genes Activities References

Tauopathies Neurofibrillary

tangles/tau

MAPT Phosphorylation

Glycosylation

Acetylation

Methylation

Ubiquitylation

SUMOylation

[113,297]

Abbreviations: PTM, posttranslational modification; CSPa, cysteine-string protein a.

S. Sharma et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions - (2018) 1-194

378

379

380

381

382

383

384

385

386

387

388

389

390

391

392

393

394

395

396

397

398

399

400

401

402

403

404

405

406

407

408

409

410

411

412

413

414

415

416

417

418

419

420

421

422

423

424

425

426

427

428

429

430

431

432

433

434

435

436

437

438

439

440

441

442

443

444

445

446

447

448

449

450

451

452

453

454

455

456

457

458

459

460

461

462

463

464

465

466

467

468

469

470

471

472

473

474

475

476

477

478

479

480

481

482

483

484

485

486

487

488

489

490

proteins. For example, abnormally hyperphosphorylated tauhas been associated with aggregates in AD and other tauopa-thies [33]. In addition, more than 40 sites of phosphorylationin tau have been described that enhance or reduce its aggre-gation propensity [33,34]. S129 is the most prominent tauphosphorylation site. The degree of tau phosphorylationdiffers in AD and FTD, which increases its propensity toaggregate and/or induce toxicity [35,36].

Although the causation and correlation between the phos-phorylation and a–synuclein (aSyn) aggregation and toxicityin Lewy bodies remain unclear, the phosphorylation state ofaSyn in Lewy bodies is well established [37,38].Approximately 90% of the phosphorylated aSyn is present inLewy bodies, a hallmark of PD, and other synucleinopathies[37,39–44]. Of the 17 potential phosphorylation sites inaSyn, S87 is the most tightly associated one withsynucleinopathies [45]. Several potential reasons for the lackof consensus among different studies may be due to in vitroand in vivo comparisons, different efficiencies of kinases, andthe balance between the phosphorylation and dephosphoryla-tion of proteins in different model systems.

Both Parkin and PTEN-induced putative kinase 1(PINK1) are mutated in PD with approximately half of theearly onset familial cases caused by Parkin [46]. Autoinhibi-tion of Parkin, an E3 ubiquitin ligase, is released on its phos-phorylation at S65 by PINK1 [47,48]. Activated Parkin

Table 2

Minimotif enzymes, activities, and substrates in familial NDs

ND(s)

Minimotifs

Enzymes

AD PS1, PS2

ALS/FTD

FTD

FTD

FTD TBK1

FTD VCP, CHMP2B, TARDBP, SQSTM1, DCTN1

HD -

PD Parkin

PD LRKK2, PINK1

PD UCH-L1

SMA UBE1, DYNC1H1

Abbreviations: AD, Alzheimer’s disease; ALS, amyotrophic lateral sclerosis; F

disease; SMA, ---.

FLA 5.5.0 DTD � TRCI168_proof

initiates a downstream ubiquitylation pathway thatregulates the quality control of proteins in PD [49].

Phosphorylation is also important for several genes with afamilial linkage to NDs. At least part of leucine-rich repeatkinase 2’s (LRKK2) pathogenicity in PD is due to its kinaseactivity and its substrate recognition likely by minimotifsthat bind its WD40 domain [50]. Ataxin1 has polyglutamine(polyQ) tracts important for the pathogenesis of spinocere-bellar ataxia (SA). Phosphorylation of S776 in Ataxin1 atan Akt phosphorylation minimotif increases cytotoxicity oflonger polyQ tracts [51,52]. Phosphorylation likely has aprotective role in ALS as a T2D phosphomimetic mutationstabilizes superoxide dismutase (SOD) in the presence ofother destabilizing pathogenic mutations [53]. Haplotypeswith this variant and a destabilizing SOD variant is onepossible explanation for variable penetrance of the patho-genic mutations.

2.1.3. LipidationFatty acid attachment to proteins anchors them to mem-

branes [54]. Some common lipidation PTMs are palmitoyla-tion, N-myristoylation, farnesylation, geranylgeranylation,GPI addition, S-diacylglycerol addition, and prenylation[54]. We do not know of a direct linkage of lipidation withproteins harboring mutations in NDs, suggesting that lipida-tion is a downstream change. However, several proteins with

Activity Substrates

Proteolysis APP

SOD1

Phosphorylation Tau

Binding SQSTM1

Kinase

HTT

Ubiquitylation a-SP22

Phosphorylation

Proteolysis Ub

Ubiquitylation

TD, frontotemporal dementia; HD, Huntington’s disease; PD, Parkinson’sQ24

� 25 July 2018 � 9:16 am � ce

491

492

493

494

495

496

497

498

499

500

501

502

503

504

505

506

507

508

509

510

511

Page 5: Minimotifs dysfunction is pervasive in neurodegenerative

7

8

9

S. Sharma et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions - (2018) 1-19 5

512

513

514

515

516

517

518

519

520

521

522

523

524

525

526

527

528

529

530

531

532

533

534

535

536

537

538

539

540

541

542

543

544

545

546

547

548

549

550

551

552

553

554

555

556

557

558

559

560

561

562

563

564

565

566

567

568

569

570

571

572

573

574

575

576

577

578

579

580

581

582

583

584

585

586

587

588

589

590

591

592

593

594

595

596

597

598

599

600

601

602

603

604

605

606

607

608

609

610

611

612

613

614

615

616

617

618

619

620

621

622

623

624

625

626

627

628

629

630

631

632

633

634

635

636

637

638

639

640

641

642

643

644

645

established roles in ND pathogenesis are covalently linked tofatty acids supporting some role. Palmitoyl acyltransferases(PATs), enzymes that catalyze protein palmitoylation recog-nize theJbxxQP minimotif (J, an aliphatic amino acid; b,a C-b branched amino acid Val, Ile, or Thr) in Huntingtin(HTT) [55–61]. ZDHHC17 is a PAT specific for neuronalproteins, including HTT. The reduced palmitoylated HTTin HD implies a reduced activity of PAT for HTT.Palmitoylated HTT has shorter polyQ tracts supporting aprotective role against HT. Similar to palmitoylation,myristoylation of HTT is also reduced in HD [62]. Onepossible explanation is that the expansion of the polyQ tractinterferes with this PTM. Palmitoylation of mutant superox-ide dismutase 1 (SOD1) and mutant cysteine-string protein amay play a role in the etiology of familial ALS and neuronalceroid lipofuscinosis, respectively [63–66].

In AD, a palmitoylated amyloid precursor protein (APP)is restricted to the Golgi, enhancing proteolytic processingthat favors amyloidogenesis. Several other substrates ofPATs associated with AD, PD, and HD are discussed [61].

Other examples of lipidation impacting NDs are a glyco-sphingolipids binding minimotif [K/H/R]x(1-4)[Y/F]x(4-5)[K/H/R], with at least one Gly at x(1-4) is present in aSyn, b-amy-loid peptide, and PrP [67,68]. Of the many gangliosides (GM),aSyn specifically binds GM1 and GM3 inhibiting fibrilformation in PD. Conversely, b-amyloid binding to GM1initiates fibril formation in AD [69,70]. PrP localization tolipid rafts stabilizes its nonpathogenic state. [L/V]x(1–5)Yx(1–5)[K/R] is a cholesterol recognition/interaction minimotifidentified in aSyn as VLYVGSK, on binding cholesterol wasaSyn aggregates in neurons [68,71]. Other types of fatty acidattachment are prenylation, geranylgeranylation, andfarnesylation. A recent review summarizes accumulatingevidence supporting a role for prenylation of small GTPasesin Ab production and secretion in AD [72].

2.1.4. Acetylation and methylationThe role of protein acetylation and methylation in NDs

was previously reviewed [73]. These PTMs are commonfor histones and can impact epigenetic inheritance, a topicaddressed later in the epigenetics section. Protein acetylationand methylation increase the surface area of, and depolarizesLys and Arg residues, a modification that generally drivesnew protein-protein interactions.

Protein acetylation is a reversible attachment of acetylgroup to the a-amino group of either the N-terminus or theε-aminogroup of theLys residues [74].Acetylation ofLys res-idues in tau inhibits its degradation and induces its aggregationin AD [75,76]. In particular, the K174Qmutant tau mimics anacetylated Lys state, leading to more severe neuronal atrophyin mice. Conversely, acetylation of a-tubulin, a PTM thatincreases its stability, is decreased in AD [73]. FTD micemodels have similar effects of acetylated tau and the K174Qmutant [32,77]. aSyn, HTT, b-amyloid, and PrP are allsubject to acetylation with either a protective or adetrimental effect on ND pathology [74,78–82].

FLA 5.5.0 DTD � TRCI168_proof

The ε-amino moiety of Lys can be mono-, di-, or tri-methylated, and Arg can be mono- or di-methylated.Methylated residues identified in tau are located in themicrotubule binding repeat region, thus, may block its in-teractions with microtubules [75]. Tandem mass spectrom-etry analysis of a human AD brain identified seven Lysresidues (K44, K163, K174, K180, K254, K267, and K290) intau that are monomethylated, and neurofibrillary tanglesare immunopositive [75]. In vitro experiments demonstratethat Qmethylation reduces deimination of Arg residues inMBP, a modification that may sensitize T-cells for autoim-mune attack of myelin in multiple sclerosis (MS) [83].

2.1.5. Nitrosylation, glutathionylation, and other oxidativePTMs

There are several types of Cys REDOX PTMs and corre-sponding minimotifs in NDs [84]. During oxidative stress,two Cys residues can oxidize creating intermolecular disul-fide bridges and protein aggregation. Some Cys thiol groupsin SOD1 are oxidized to sulfenic, sulfinic, and sulfonicacids, decreasing the stability of the protein and renderingit prone to aggregation in ALS patients [63,85–88].Similarly, S-glutathionylation of Cys residues in SOD1induces its aggregation in ALS [89,90].

Aberrant protein S-nitrosylation in several NDs,including ALS, AD, and PD, has been previously reviewed[88,91]. A global analysis of mouse brain identified 31nitro-tyrosine PTMs, more than half of which have beenimplicated in PD, AD, and other NDs [92–94]. Theconsensus motif for nitric oxide reaction with proteins is[K/R/H/D/E]C[D/E], where the Cys residues is covalentlyattached to nitric oxide [95,96]. Protein S-nitrosylation caninduce protein misfolding and aggregation [95,97].Aberrant S-nitrosylation of many proteins (parkin, PDI,DNM1L, CDK5, PRRX2, GAPDH, PTEN, AKT1, MAPK,IKBKP, and XIAP) may contribute to neurodegenerationthrough multiple pathways [91]. For example, aberrant S-ni-trosylation of PDI decreases its enzymatic activity, therebyinhibiting its neuroprotective functions [91].

2.1.6. UbiquitylationMany misfolded and damaged Qproteins are targeted for

ATP-dependent proteolysis by conjugation with ubiquitin,an 8.5 kDa protein. The role of ubiquitylation in NDs waspreviously summarized [98]. Proteins that aggregate inNDs tend to be ubiquitylated, likely for initiating targetedprotein degradation. In the cerebral cortex Qof patients withAD, the cerebral spinal fluid of patients with CJD, andaSyn in patients with PD, ubiquitylation of neurofibrillarytangles is increased [99–102]. Tau, the protein thatoligomerizes in these tangles, is ubiquitylated at K254 inAD, and the ubiquitylated tau may be blocked by Lysacetylation in FTD [32,76]. A PolyQ tract in Ataxin-1 in-duces its aggregation in the nucleus and leads to ubiquity-lated SA type 1 (SCA-1), inducing cytotoxicity in SA[103]. Ubiquitylated polyQ tracts in HD and other triplet

� 25 July 2018 � 9:16 am � ce

Page 6: Minimotifs dysfunction is pervasive in neurodegenerative

10

S. Sharma et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions - (2018) 1-196

646

647

648

649

650

651

652

653

654

655

656

657

658

659

660

661

662

663

664

665

666

667

668

669

670

671

672

673

674

675

676

677

678

679

680

681

682

683

684

685

686

687

688

689

690

691

692

693

694

695

696

697

698

699

700

701

702

703

704

705

706

707

708

709

710

711

712

713

714

715

716

717

718

719

720

721

722

723

724

725

726

727

728

729

730

731

732

733

734

735

736

737

738

739

740

741

742

743

744

745

746

747

748

749

750

751

752

753

754

755

756

757

758

759

760

761

762

763

764

765

766

767

768

769

770

771

772

773

774

775

776

777

778

779

repeat NDs enhance intranuclear protein aggregation in neu-rons [104–106]. Additional PTMs in the polyQ tracts in 12proteins involved in the pathology of 9 NDs are known[107]. SCF E3 ubiquitin ligase binds survival motor neuronprotein through a phosphodegron signal, DSGxx[S/T],where the Ser is phosphorylated [108]. This suggests thatoligomerization of the mutant survival motor neuron poten-tially sequesters the degradation signal and stabilizes sur-vival motor neuron, a potential pathogenic pathway inspinal muscular atrophy [108,109].

2.1.7. SumoylationSmall Ubiquitin-like Modifiers (SUMOs) are ubiquitin-

like proteins covalently attached to other proteins atJKXE/D minimotifs. There are four SUMO genes in hu-mans [110]. SUMO normally functions in protein stability,nuclear-cytosolic transport, and transcriptional regulation.Many key proteins in ND pathogenesis are sumoylated andimplicated in disease mechanisms [111–113]. In AD,sumoylation of APP decreases Ab production, andsumoylation stabilizes tau by inhibiting itsphosphorylation and ubiquitylation [113–116]. Similarly,in PD, sumoylation of aSyn inhibits its aggregation[117,118]. The pathogenic fragment of HTT is sumoylatedin HD, and SOD aggregation in ALS is also effected [119–122]. Pathogenic mutation of sumoylated residues invalosin-containing protein/p97 inhibits its translocation tothe nucleus, the formation of stress granules, reduction ofhexamer formation, and eventually, inhibition of its clear-ance. These mutants are prevalent in FTDs [123].

2.1.8. CitrullinationCitrullination is an irreversible PTM type of minimotif

[124]. Peptidylarginine deiminases deiminate Arg residuesin proteins converting this amino acid to citrulline. Becausearthritic patients have anti-citrullinated protein antibodies,citrullination may be related to some of the neuroinflamma-tory aspects of NDs. Abnormal protein citrullination isknown for AD, PD, including Lewy bodies, and in prion dis-eases [125–127]. Autoimmune attack of citrullinatedproteins may be prevalent in ND neuroinflammation[128,129]. Although MS is not considered a ND,citrullination of the major myelin component, MBP mayplay a role in the autoimmune attack of myelin in MS[130]. Deimination depolarizes MBP and is a modificationthat may trigger the demyelination of axons in MS [131].

2.1.9. EndoproteolysisProteolysis is central to the degradation of misfolded pro-

teins by the ubiquitin-proteasome system and autophagy,both driven through minimotifs and central to NDs [132].The inherent role of endoproteolysis in neurodegenerationwas founded in the familial genetics of APP processing tob-amyloid and was recently reviewed [133]. In early onsetAD, b-secretase and g-secretase cleave APP-producingamyloidogenic Ab40 or Ab42 [27,134,135]. Familial

FLA 5.5.0 DTD � TRCI168_proof

mutations in the protease processing minimotif sites inAPP cause overproduction of amyloidogenic peptidefragments.

Other NDs have proteolysis minimotifs as well. In pa-tients with PD, familial mutations were identified inUCHL1, a protease that cleaves ubiquitin and colocalizeswith Lewy bodies. Cleavage of the furin prohormone con-vertase cleavage Qsite (KGIQKREA) in putative type-II sin-gle-spanning transmembrane precursor protein (BRI)yields a small C-terminal amyloidogenic peptide fragment[136]. These fragments are present in the amyloid fibrils ofpatients with Familial British dementia [137–139]. Severalproteases cleave HTT at minimotifs producing peptideaggregation and neurotoxic peptides [140–144].

2.2. Trafficking and autophagy minimotifs inneurodegenerative diseases

Minimotifs are important in protein trafficking to andfrom organelles and are relevant to several NDs[1,145,146]. Defects in endocytic and synaptic vesicletrafficking, Golgi trafficking, retrograde transport, andlysosomal autophagy are apparent in PD and AD[145,147,148]. Trafficking minimotifs are essential formost protein trafficking events, have been previouslyreviewed, and a few typical examples are presented [148–151]. LRKK2 is a kinase/GTPase with mutations inpatients with familial PD [152]. LRRK2 has a WD40 inter-action domain that binds to synaptic vesicles and severalLRRK2 interactors function in vesicular trafficking[50,153]. Through phosphorylation of S75 in endophilin,LRKK2 plays a role in synaptic vesicle endocytosis inDrosophila [154]. LRKK2 is also involved in retrogradetrafficking through a functional association with VPS35,another protein mutated in familial PD.

The role of trafficking of APP in AD pathogenesis hasrecently been reviewed [135]. A KFERQ minimotif at theend of APP binds SCG10 and is essential for traffickingAPP to lysosomes for degradation [155,156]. The KDELreceptor binds ER resident proteins containing the KDELminimotif and returns them to the endoplasmic reticulum[157]. The receptor is redistributed to lysosomes and stimu-lates autophagy, when aberrant SOD in ALS, aSyn in PD,and HTT in HD are expressed [158].

The cytoplasmic domain of APP contains a YxNPxYsequence, an internalization minimotif that traffics APP toendosomes [159,160]. The same minimotif in APP bindsto adaptor proteins, including LDL-Rs, FE65, X11,SNX17, and Dab2, which regulate APP trafficking andthereby regulates b-amyloid production [159].

Mutant ataxin-1 has a nuclear localization signal, whichis required to mediate toxicity of variants with long polyQtracts in SAs [51,161]. Mutations in the VxPx. minimotifin rhodopsin cause autosomal dominant retinitispigmentosa, a progressive ND of photoreceptor neurons[162]. This C-terminal minimotif is essential for trafficking

� 25 July 2018 � 9:16 am � ce

Page 7: Minimotifs dysfunction is pervasive in neurodegenerative

Q11

S. Sharma et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions - (2018) 1-19 7

780

781

782

783

784

785

786

787

788

789

790

791

792

793

794

795

796

797

798

799

800

801

802

803

804

805

806

807

808

809

810

811

812

813

814

815

816

817

818

819

820

821

822

823

824

825

826

827

828

829

830

831

832

833

834

835

836

837

838

839

840

841

842

843

844

845

846

847

848

849

850

851

852

853

854

855

856

857

858

859

860

861

862

863

864

865

866

867

868

869

870

871

872

873

874

875

876

877

878

879

880

881

882

of rhodopsin from the trans-Golgi-network [163]. This mini-motif also binds to ADP-ribosylation factor 4, a rhodopsintransport carrier, regulating the otherwise bulk flow of pro-teins to photoreceptor rod outer segments.

Chaperone-mediated autophagy, in addition to ubiquityla-tion, is another mechanism that maintains protein quality con-trol in NDs [164]. The substrates of autophagy contain aKFERQ-like minimotif that is exposed potentially only aftera PTM-induced conformational change. These minimotifsinteractwith the chaperone complex and are translocated to ly-sosomes via lysosomal-associatedmembrane protein 2A, a re-ceptor that triggers autophagy. AVKKDQ minimotif in aSyntargets it for autophagy [165,166]. aSyn A30P and A53Tmutants may cause familial PD by interacting withlysosomal-associated membrane protein 2A. These mutantproteins fail to translocate into the lysosomal lumen, effi-ciently stalling the process of autophagy.

Autophagy may be a step in the pathology of PD and HD.Autophagy-targeting minimotifs (QVEVK, KDRVQ) in tauare important in a neuronal cell model of tauopathies [167].On binding the chaperone complex, Cathepsin L, a lysosomalcysteine protease cleaves mutant tau proteins producing pep-tide fragments that aggregate [167]. Amore detailed overviewof autophagy enumerates eight proteins containing 17confirmed and putative autophagy-targeting minimotifs inproteins involved in PD etiology [168]. Mutant KDRVN andNEIKV minimotifs in HTT are recognized by the autophagymachinery, but an extended C-terminal polyQ track in HTTdelays targeting to the lysosomes. This may result in severeHTT aggregation in the neurons of patients with HD [169].

883

884

885

886

887

888

889

890

891

892

893

894

895

896

897

898

899

900

901

902

903

904

905

906

907

908

909

910

911

912

913

2.3. Binding minimotifs in neurodegenerative diseases

One of the most important activities of minimotifs is forproteins to bind other molecules, which is central to manycellular pathways and processes. Interestingly, many typesof bindingmotifs propagate neuronal atrophy inNDs. A link-age analysis of a Turkish kindred identified an autosomalrecessive variant encoding a D458Vmutation in the C-termi-nal PDZ binding motif of SANS protein in patients with theatypical Usher syndrome [170,171]. Through SH3 bindingminimotif, RTPPKSP, tau binds the SH3 domains of Fynand Src nonreceptor tyrosine kinases, which alsophosphorylate tau [172,173]. Because hyperphosphorylatedtau is a hallmark of AD [174]. MED25 binds SH3 domainsof Abelson family protein kinases. The A335V mutation inthe proline-rich SH3 binding motif of MED25 decreasesbinding specificity imparting interactions with a broaderrange of nonphysiological proteins with SH3 domains. Thismutation causes Charcot-Marie-Tooth disease 2B2 [175]. AYENPTY minimotif in APP interacts with PTB domains ofMint proteins trafficking and processing APP [176–178].Ataxin has a RxxSxP 14-3-3 binding minimotif thatenhances the toxicity of its polyQ tracts in SA [179].

The GxxxGxxxG, glycine zipper minimotif binds choles-terol and has an established role in oligomerization of pro-

FLA 5.5.0 DTD � TRCI168_proof

teins, including Ab and PrP [180–182]. Cholesterolbinding to APP may contribute to amyloidogenesis andAD [180]. In the postsynaptic terminal, b-amyloid recruitsPTEN, a lipid phosphatase in a PDZ domain-dependentmanner inducing a malfunctional state of amyloids [183].A PTEN mutant lacking its PDZ domain protects againstamyloid toxicity.

2.4. Minimotif cooperativity

Minimotif cooperativity is perhaps one of the most under-studied aspects of minimotif function and is likely an impor-tant factor for understanding molecular dysregulation inNDs. Many proteins, including those associated with NDs,have more than five minimotifs, and there are now numerousexamples of how one minimotif can induce or create a newminimotif, or compete with other minimotif targets for thesame site. These often impact a protein activity or cell pro-cess. Several examples of minimotif cooperativity arementioned in the other sections, and a few are highlightedhere. In PTM minimotifs, certain residues either competefor a particular PTM or induce further modifications. Forexample, phosphorylation often precedes modificationssuch as sumoylation and ubiquitylation [38]. In patientswith AD, tau phosphorylation follows an initial glycosyla-tion modification [30,184]. When MARK2 phosphorylatestau, it is no longer recognized by E3 ligase, preventing itsdegradation [185]. Methylation minimotifs in tau are posi-tioned to engage in cross talk with phosphorylation [75].

3. Minimotifs in aggregates

Protein aggregation is common in the etiology of mostNDs [186]. b-Amyloid and tau are the major constituentsof neuritic plaques and neurofibrillary tangles, respectively.Lewy bodies are intracellular aggregates of aSyn in PD andother synucleinopathies. Proteins with extended polyQ re-peats aggregate into nuclear and cytosolic inclusions inHD, SA, and dentatorubral-pallidoluysian atrophy. A hall-mark of ALS is cytoplasmic inclusions of SOD1.

Although abnormal protein aggregation in neurodegener-ation is well established, the generalized role of minimotifsin aggregation was not previously emphasized. There areseveral genes with familial inheritance in NDs that encodeminimotif target enzymes or their substrates. These genes,minimotifs, and their activities are listed in Table 2. b-Am-yloid, the central component of plaques, is ubiquitylatedand phosphorylated, which regulates the stability of plaques[187–189]. Most tauopathies have the common molecularabnormality of hyperphosphorylation of tau, whichaggregates into neurotoxic inclusions [190]. Furthermore,there are at least a dozen other PTMs that modulate tau ag-gregation [116,191]. Lewy bodies are ubiquitylated,sumoylated, phosphorylated, nitrosylated, and also have aunique PTM created by a dopamine radical adduct[117,192,193].

� 25 July 2018 � 9:16 am � ce

Page 8: Minimotifs dysfunction is pervasive in neurodegenerative

Q12

S. Sharma et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions - (2018) 1-198

914

915

916

917

918

919

920

921

922

923

924

925

926

927

928

929

930

931

932

933

934

935

936

937

938

939

940

941

942

943

944

945

946

947

948

949

950

951

952

953

954

955

956

957

958

959

960

961

962

963

964

965

966

967

968

969

970

971

972

973

974

975

976

977

978

979

980

981

982

983

984

985

986

987

988

989

990

991

992

993

994

995

996

997

At least 48 PTMs of protein have been identified in NDs,and other neurotoxic proteins with long polyQ tracts havemany PTMs [194,195]. Because minimotifs are located onthe surfaces of all proteins and cover almost the entiresurface of many proteins [196–199], it is not surprisingthat perturbing minimotifs results in abnormal neurotoxicaggregation. Given their prevalence in neurotoxicaggregate formation and stability, a betting understandingof how minimotifs cooperate to induce and inhibitneurotoxic aggregates would provide a betterunderstanding of ND etiology and open the door for newtypes of therapeutic intervention. As such, minimotifmimetics should be considered as targets for reducingaggregation in NDs.

13

998

999

1000

1001

1002

1003

1004

1005

1006

1007

1008

1009

1010

1011

1012

1013

1014

1015

1016

1017

1018

4. Genetics and epigenetics of minimotifs in NDs

Over the past decade, genome-wide association studies(GWASs) have advanced our understanding of the geneticbasis of human disease [200]. Similar to other disorders,NDs have a familial component with rare variants of largeeffect and a more prevalent sporadic component withmany common variants of small effect. Given that minimo-tifs are located at the interface between proteins and areimportant constituents of cellular networks, it is no surprisethat several of these motifs have been highlighted in GWASsand in the biology of key cell types such as neurons, astro-cytes, and microglia. Some of the earliest advances in ge-netics were derived from genetic linkage and positionalcloning in NDs and have led to newer studies relying ongenome-wide quantitative trait analyses such as biomarkersor endophenotypes [201].

1019

1020

1021

1022

1023

1024

1025

1026

1027

1028

1029

1030

1031

1032

1033

1034

1035

1036

1037

1038

1039

1040

1041

1042

1043

1044

1045

1046

1047

4.1. Genetics

Through the combination of high-throughput genotypingplatforms and next-generation sequencing technologies,disease-causing genetic variants and genetic factors necessaryfor protection against disease have been discovered at an un-precedented rate. While such technologies have transformedmonogenic disease research, several challenges continue tohamper the discovery of actionable genomic variants in com-plex disorders. For example, variants of unknown significanceand an abundance of neutral variants or variants with modesteffect size continue to be produced at a staggering rate. Assuch, leveraging next-generation sequencing for discoveryhas been contingent on carefully constructed studies of largesample sizes and judicious selection of participants with accu-rate and detailed phenotyping. Here, we focus on AD and PDas exemplars of NDs, and we also discuss a pervasive role ofgenetic variants in minimotifs.

4.1.1. Alzheimer’s diseaseAD is a common neurodegenerative disease and a leading

cause of dementia with progressive loss of memory,problem-solving skills, and communication. Due to recent

FLA 5.5.0 DTD � TRCI168_proof

improvements in life expectancy, AD is predicted to affect1 in 85 people globally by 2050 [202]. As a heterogeneousdisease, AD is caused by a combination of environmentaland genetic factors and current estimates of AD heritabilitylie between 60-80% [203]. Late-onset AD accounts for morethan 95% of all AD cases and is caused by a complex under-lying genetic architecture. To date, over 40 GWASs andmeta-analyses have identified a few hundred genes and sin-gle nucleotide polymorphisms; these include variants inapolipoprotein E (APOE) among other notable gene candi-dates [204]. In contrast to late-onset AD, early onset AD iscaused by highly penetrant variants located primarily inthree genes, APP (located at 21q21.2), presenilin 1(PSEN1, located at 14q24.3), and presenilin 2 (PSEN2,located at 1q42.13) [202]. Structural variants are associatedwith complex neurological traits; for example, duplicationof APP causes early-onset AD with cerebral amyloid angi-opathy [205,206]. To empower current association studies,new attempts at incorporating quantitative endophenotypessuch as age at onset analysis and expression quantitativetrait loci are being conducted [207].

Minimotif activities are prevalent in genes associatedwith AD. On examining disease genes with penetrant vari-ants, we identified at least 11 activities that could be associ-ated with disease: proteolysis, sumoylation, glycosylation,lipidation, acetylation, methylation, oxidative PTM, ubiqui-tylation, citrullination, trafficking, and binding (Tables 1 and2). Intriguingly, more than half of all APP mutations arelocated at the secretase proteolytic cleavage sites or thetransmembrane domain, a region on exon 16/17 (www.molgen.ua.ac.be/ADMutations). In addition, loss-of-function AD Qanimal and cellular models suggest that suchminimotif activities are indeed perturbed functionally. Assuch, we predict that coding and structural variants thatimpact minimotif function will be key nodes in ND [208].

4.1.2. Parkinson’s diseaseAs the second most common neurodegenerative disorder

after AD, PD results from the loss of dopaminergic neuronsin the midbrain and the accumulation and aggregation ofaSyn in Lewy bodies [209,210]. While rare before the ageof 60 years, up to 4% of the population at the age of80 years has PD [211]. Similar to AD, the genetic architec-ture of PD is complex, and only 5-10% of all patients sufferfrom an apparent monogenic form of PD [211]. These rareand penetrant variants are located within 19 disease-causing genes such as aSyn (SNCA, located at 4q22.1),LRRK2 (located at 12q12), and acid b-glucosidase (GBA,located at 1q22) and segregate with the disease in an auto-somal dominant or recessive pattern [212]. GWASs haverecently identified more than 40 risk loci, including candi-date genes that LRRK2 phosphorylates [213].

In a similar minimotif analysis of PD, we note that 11 activ-ities are potentially impacted. These include the following: pro-teolysis, sumoylation, glycosylation, lipidation, acetylation,methylation, oxidative PTM, ubiquitylation, phosphorylation,

� 25 July 2018 � 9:16 am � ce

Page 9: Minimotifs dysfunction is pervasive in neurodegenerative

Q14

S. Sharma et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions - (2018) 1-19 9

1048

1049

1050

1051

1052

1053

1054

1055

1056

1057

1058

1059

1060

1061

1062

1063

1064

1065

1066

1067

1068

1069

1070

1071

1072

1073

1074

1075

1076

1077

1078

1079

1080

1081

1082

1083

1084

1085

1086

1087

1088

1089

1090

1091

1092

1093

1094

1095

1096

1097

1098

1099

1100

1101

1102

1103

1104

1105

1106

1107

1108

1109

1110

1111

1112

1113

1114

1115

1116

1117

1118

1119

1120

1121

1122

1123

1124

1125

1126

1127

1128

1129

1130

1131

1132

1133

1134

1135

1136

1137

1138

1139

1140

1141

1142

1143

1144

1145

1146

1147

1148

1149

1150

1151

1152

1153

1154

1155

1156

1157

1158

1159

1160

1161

1162

1163

1164

1165

1166

1167

1168

1169

1170

1171

1172

1173

1174

1175

1176

1177

1178

1179

1180

1181

citrullination, and trafficking (Tables 1 and 2). To verify therelevance of these predictions, we examined how familial mu-tations inLRRK2might affectminimotif activities (Table 2).Onfocusing on phosphorylation, we noted that disease-associatedmutations, Y1699C and I2020T (nonphosphorylated residues),disrupted the phosphorylation of constitutively phosphorylatedresidues at the amino terminal, resulting in attenuated LRRK2function [214]. These findings, and additional LOF and GOFmodels, suggest that detailed analyses of minimotif functioncan identify feedback control mechanisms and novel networksthat drive PD biological processes [215].

4.1.3. Microglia and NDsMicroglia are the highly specialized macrophages of the

central nervous system, accounting for 10–15% of all cells inthe adult brain [216]. Significant advances have been madein understanding the roles of microglia in the developmentof the brain, such as in neurogenesis, synaptic pruning, sur-veillance, and homeostasis [217]. In contrast, defining therole of microglia in central nervous system disorders hasproven to be more difficult. Given that microglial activationand neuroinflammatory processes play a critical role in thepathogenesis of NDs, biological components linked to mi-croglial biology have been associated with AD etiologythrough GWAS; these include the following: TREM2 (trig-gering receptor expressed on myeloid cells 2), CD33, CR1(complement receptor 1), ABCA7 (ATP-binding cassette,sub-family A, member 7), SHIP1 (also known as INPP5D,inositol polyphosphate-5-phosphatase D), APOE, CLU(clusterin), CD2AP (CD2-associated protein), and EPHA1(EPH receptor A1) [218–222].

Complementing the human genetic studies, knockoutmice models of genes from GWAS have allowed forcause-effect in vivo investigations into disease pathogenesis.As expected, a fraction of these genes is implicated in theregulation Ab accumulation. For example, ApoE- andCLU-deficient APP transgenic mice exhibit earlier andmore extensive Ab deposition compared with control mice[223]. In addition, CD33 inactivation in App/Psen1 micereduced Ab accumulation and plaque burden, whereasAbca7 deficiency in App/Psen1 and TgCRND8mice acceler-ated Ab generation [224–226]. Importantly, variants ofTREM2, an immunoglobulin-like cell-surface receptor spe-cifically expressed in brain microglia, confer a 2- to 4-foldincreased risk for AD [221]. However, exactly how Trem2variants confer AD risk is still under investigation. Thusfar, the most consistent and striking observation is a strongdecrease in microgliosis surrounding Ab plaques of AD inTrem2 haploinsufficient and Trem2 deficient mice; a similarimpairment in microgliosis has also been reported in mousemodels of prion disease, stroke, and MS, suggesting a crit-ical role for TREM2 in supporting microgliosis in responseto pathology in the central nervous system.

A loss-of-function mutation in the lipid sensing function,a minimotif activity of Trem2 deficient mice may be the un-derlying mechanism for the loss of microglia proliferation

FLA 5.5.0 DTD � TRCI168_proof

and microglial response to Ab plaques or reduced infiltrationof peripheral macrophages [227,228]. In addition, Trem2deficiency leads to exacerbated aggregation of tau in amouse model of tauopathy [229]. These findings demon-strate that TREM2 has complex multiple roles in regulatingAb and tau pathologies that may reflect its distinct functionsat different stages in AD pathology [230]. These findingsalso highlight how minimotif activities are present in severalproteins relevant to microglial biology.

Minimotifs are a source of functional genetic variationand are important targets in natural selection and evolution[231]. Given their prevalence in NDs, we must considerthat common NDs may include a cumulative compositionof many minimotif variants that cause network dysfunction.This could provide an explanation for missing heritability inNDs. One exciting possibility is that minimotifs maycontribute to the genetic risk of neurodegeneration; however,this will require additional study.

4.2. Epigenetics

Epigenetics refers to inheritance that is notmediated throughDNA sequence but through covalentmodifications or noncova-lent DNA interactions. A number of recent reviews highlightthe emerging role of epigenetics in normal brain function andneurodegeneration [232,233]. Major mechanisms ofepigenetics are DNA methylation and PTM of histones/nucleosomes, which is often referred to as the histone code[234]. Minimotifs are the foundation of the histone code. En-zymes that covalently modify minimotifs, including those inhistones, are central to AD, PD, ALS, HD, and other polyQtrack disorders summarized in recent reviews [235–240].

The role of minimotifs in ND epigenetics is of general in-terest for ND beyond disease etiology. One emerging area ofinterest is drugging the enzymes that modify protein compo-nents of nucleosomes. Considering the broad role of epige-netics in NDs, there are several drugs such as histonedeacetylase inhibitors that are being tested inpreclinical inves-tigation and are in different phases of clinical trials [233].

Epigenetics may also explain at least part of the ubiqui-tous “missing heritability” for common diseases such asthe major NDs. Testing the role of epigenetics in PD mayprove difficult without considering the effects of minimotifs.This is because our group has previously reported histonecode variability in the population [7]. Approximately 4%of the minimotifs in histone tails had common alleles in hu-mans that were loss-of-function for the PTM. This suggeststhat the histone code may be wired differently among peo-ple, inferring differing epigenetic responses to the environ-ment [7]. This may be critical to account for in any studythat attempts to uncover the epigenetic basis of NDs.

5. Minimotifs as therapeutic targets for NDs

The Therapeutics Peptide Database has no FDA-approved drugs for treating NDs [241]. Despite this lack of

� 25 July 2018 � 9:16 am � ce

Page 10: Minimotifs dysfunction is pervasive in neurodegenerative

Q15

S. Sharma et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions - (2018) 1-1910

1182

1183

1184

1185

1186

1187

1188

1189

1190

1191

1192

1193

1194

1195

1196

1197

1198

1199

1200

1201

1202

1203

1204

1205

1206

1207

1208

1209

1210

1211

1212

1213

1214

1215

1216

1217

1218

1219

1220

1221

1222

1223

1224

1225

1226

1227

1228

1229

1230

1231

1232

1233

1234

1235

1236

1237

1238

1239

1240

1241

1242

1243

1244

1245

1246

1247

1248

1249

1250

1251

1252

1253

1254

1255

1256

1257

1258

1259

1260

1261

1262

1263

1264

1265

1266

1267

1268

1269

1270

1271

1272

1273

1274

1275

1276

1277

1278

1279

1280

1281

1282

1283

1284

1285

1286

1287

1288

1289

1290

1291

1292

1293

1294

1295

1296

1297

1298

1299

1300

1301

1302

1303

1304

1305

1306

1307

1308

1309

1310

1311

1312

1313

1314

1315

success, the investigation of minimotif-directed therapeuticshas been and remains of high interest and potential. Thera-peutic interventions based on minimotifs have been dis-cussed in detail for AD, PD, HD, and CJD [242–249].Since APP was one of the first genes connected withfamilial AD and the aggregation of Ab is a predominatefeature of late-onset AD, there has been much interest in in-hibiting Ab production. Inhibitors of a, b, and g-secretasethat inhibit APP processing, Ab production, and aggregationis still under clinical study as was recently reviewed [250].

Several other minimotif-directed therapeutics are beinginvestigated for reducing aggregation of neurotoxic proteins.One central therapeutic approach, although not directed to-ward minimotifs, is to lower levels of the neurotoxic proteinby antisense oligonucleotide therapy [251,252]. Theubiquitin-proteasome system is strongly associated withNDs and is a target for new therapies with compounds thatstimulate proteasome-mediated aggregate degradation ortherapeutics that target specific proteins for proteome degra-dation [98,253]. Aggregation-blocking peptides such asSNWKWWPGIFD, a polyQ-binding peptide against HTT;GAVVT, a aSyn self-aggregation blocking peptide againstPD; and a NPxY. PTB domain-interacting endocyticminimotif-blocking peptide against AD are also being tested[254–256]. Glycosaminoglycans mimetics are decoyinhibitors of covalent glycosaminoglycan attachment toAb, blocking its aggregation [257]. Polyanions such as pen-tosane polysulfate interferewith aggregation of glycosylatedPrP have shown promise in a preclinicalmodel [258]. Severalkinase inhibitors block GSK3b, CDK5, and other tau kinasesto reduce tau phosphorylation, and hence their aggregationsare being investigated for AD and tauopathies [33].

Given the role of neuroinflammation and autophagy inAb removal, drugs that enhance amyloid degradation areof growing interest. Many analogs and approaches to targetautophagy machinery were previously reviewed [132,169].Rapamycin is a FDA-approved drug that inhibits the kinaseactivity of mTOR and improves memory function in modelorganisms. Preclinical studies of mice show that rapamycinreduces Ab and tau-associated pathologies by increasingautophagy [255,256,259]. Although not related tominimotifs, lithium also induces autophagy and clearanceof HTT, synuclein, and PrP [258].

Three histone deacetylase inhibitors were approved bythe FDA for treating various types of cancer [259]. SinceNDs have an epigenetic component (addressed earlier), his-tone deacetylase inhibitors and other drugs targeting epige-netic modifications may be useful for treating NDs. This firsttest of hypothesis is currently under investigation with clin-ical trials on ORY-2001 for treating AD. ORY-2001 inhibitshistone methylation and reduces cognitive impairment andneuroinflammation in rodent model.

Althoughminimotif-targeted drugs are not a major class oftherapeutics, as we learn more about minimotifs, we hypothe-size that their broad influence on disease will likely becomemore evident and become useful for treatment of NDs.

FLA 5.5.0 DTD � TRCI168_proof

6. Conclusion

Minimotifs are short peptide sequences that encode a mo-lecular function. Their role in cancer and infectious diseaseshas been previously established [9–11]. In this review, wesummarize the evidence supporting minimotifs as keyregulators of ND pathogenesis and describe numerousexamples of causative cases in familial NDs.

Collectively, numerous studies support centrality of mini-motifs to NDs, much of which was previously reviewed[33,111,119,125,131,192,253,260–264]. Our observationof the generalized minimotif pervasiveness in NDs raisesseveral interesting questions. Particularly, why areminimotifs so prevalent in NDs, and can this observationteach us anything more general about ND etiology?

We consider that answers to these questions may arisefrom the role of minimotifs in protein and larger networks.NDs have an additional layer of complexity when comparedwith other human diseases. Many diseases are founded inphysiological dysfunction of the cellular network that man-ifests as the emergent network properties of symptoms andeven death. This network dysfunction is rooted in dysfunc-tion of the molecular network with cells where minimotifshave an important role. However, networks are more com-plex in NDs because the symptoms arise from emergentproperties of dementia, loss of motor control, and othermental health symptoms. These symptoms are due to break-down of an additional network, the neuronal network.Cognitive states are considered emergent properties of anetwork [265].

Why are minimotifs a point of vulnerability? Thus far,about a million minimotifs have been experimentally veri-fied with large growth anticipated [4]. Minimotifs are thekey determinants that enable proteins work together in anetwork as a basis for emergent properties in the cell suchas neurotransmission and neuroinflammation. Most complexnetworks, including biological networks and the minimotif/protein network, have topologies of cliquish small world net-works and have the property of robustness.

Robustness protects the system against immediate shockthrough adaptive mechanism engineered over time throughselection and evolution. Those mutations that target key vul-nerabilities in the network cause failure and prenatallethality.

While the protein/minimotif, cellular, and neuronal net-works are evolutionarily tuned for robustness, like anynetwork, they can fail, the failures of interest herein beingNDs and death. The study of ND disease etiology hasinformed us of two general categories of disease failure, fa-milial, and sporadic. Familial ND network failure may arisefrom hub protein dysfunction [266,267]. Hub proteins aregenerally highly enriched with minimotifs, and our reviewconsolidates many types of minimotifs involved in familialNDs. Hub nodes can collapse an entire network because offunctional overload and ability to redistribute informationflow [268].

� 25 July 2018 � 9:16 am � ce

Page 11: Minimotifs dysfunction is pervasive in neurodegenerative

web4C=FPO

web4C=FPO

Fig. 1. General Q18functions of minimotifs in NDs. Minimotifs perform multiple functions in NDs. Shown here are a selected functions or cell processes of

minimotifs in NDs. Abbreviations: AD, Alzheimer’s disease; ALS, amyotrophic lateral sclerosis; CJ, Creutzfeldt-Jakob; FTD, frontotemporal dementia;

HD, Huntington’s disease; NCL, neuronal ceroid lipofuscinosis; PD, Parkinson’s disease; RP, retinitis pigmentosa; SBMA, spinal and bulbar muscular atrophy;

SA, spinocerebellar ataxia.

web4C=FPO

web4C=FPO

Fig. 2. Minimotifs in neurotoxic protein aggregation. The different types of

aggregates in NDs are shown. The types of minimotifs for each aggregate

are binding (B), trafficking (T), and posttranslational modification (M).Q19 Ab-

breviations: AD, Alzheimer’s disease; ALS, amyotrophic lateral sclerosis;

CJ, Creutzfeldt-Jakob; FTD, frontotemporal dementia; HD, Huntington’s

disease; NCL, neuronal ceroid lipofuscinosis; PD, Parkinson’s disease;

RP, retinitis pigmentosa; SBMA, spinal and bulbar muscular atrophy; SA,

spinocerebellar ataxia.

FLA 5.5.0 DTD � TRCI168_proof

S. Sharma et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions - (2018) 1-19 11

1316

1317

1318

1319

1320

1321

1322

1323

1324

1325

1326

1327

1328

1329

1330

1331

1332

1333

1334

1335

1336

1337

1338

1339

1340

1341

1342

1343

1344

1345

1346

1347

1348

1349

1350

1351

1352

1353

1354

1355

1356

1357

1358

1359

1360

1361

1362

1363

1364

1365

1366

1367

1368

1369

1370

1371

1372

1373

1374

1375

1376

1377

1378

1379

1380

1381

1382

1383

1384

1385

1386

1387

1388

1389

1390

1391

1392

1393

1394

1395

1396

1397

1398

1399

1400

1401

1402

1403

1404

1405

1406

1407

1408

1409

1410

1411

1412

1413

1414

1415

1416

1417

1418

1419

1420

1421

1422

1423

1424

1425

1426

1427

1428

1429

1430

1431

1432

1433

1434

1435

1436

1437

1438

1439

1440

1441

1442

1443

1444

1445

1446

1447

1448

1449

In sporadic NDs, GWAS studies have helped resolve anetiology of the culmination of many genetic variants of smalleffect size. In this scenario, we suggest that this networkdysfunction arises from a dynamic redistribution of flowthrough the network [267]. The genetic risk score is a mea-sure of reaching a mutational burden threshold and is consis-tent with this model. While many of the variants are innoncoding regions, disruption of key minimotifs likely con-tributes to the network dysfunction. Furthermore, the birth ofbillions of people provides a screen to identify how multiplevariants can contribute to breaking network robustness man-ifested as NDs. This could also explain the higher prevalenceof sporadic disease in general.

The role of minimotifs in network dysfunction may alsoprovide an explanation for the slow progression of NDs,often over decades. ND can be considered a cascading fail-ure of networks where several models have been proposed.For late-onset AD, the uncontrolled neuroinflammation hy-pothesis proposes a feedback cycle that leads to shutdownof microglial phagocytosis of amyloid plaques [269–271].This is an example of a cascading failure of a networkwhere the disease manifest once a key threshold isbreached with overload of a subnet [272]. Minimotifs areinvolved in immune responses and autophagy and are likelypart of the network dysfunction [273].

� 25 July 2018 � 9:16 am � ce

Page 12: Minimotifs dysfunction is pervasive in neurodegenerative

Q17

Q16

20

21

S. Sharma et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions - (2018) 1-1912

1450

1451

1452

1453

1454

1455

1456

1457

1458

1459

1460

1461

1462

1463

1464

1465

1466

1467

1468

1469

1470

1471

1472

1473

1474

1475

1476

1477

1478

1479

1480

1481

1482

1483

1484

1485

1486

1487

1488

1489

1490

1491

1492

1493

1494

1495

1496

1497

1498

1499

1500

1501

1502

1503

1504

1505

1506

1507

1508

1509

1510

1511

1512

1513

1514

1515

1516

1517

1518

1519

1520

1521

1522

1523

1524

1525

1526

1527

1528

1529

1530

1531

1532

1533

1534

1535

1536

1537

1538

1539

1540

1541

1542

1543

1544

1545

1546

1547

1548

1549

1550

In conclusion, our summarization of NDs shows a perva-sive role for many different types of minimotifs in familialmutations, aggregation, the histone code, epigenetics, andbiomarkers. Minimotifs are likely a key part of networkdysfunction in NDs.

Acknowledgments

The National Institutes of Health grants R15GM107983,R01GM079689, R01LM010101, R21AI116411, andP20GM121325 was provided to M.R.S., P20GM109025and NV Governor’s Office of Economic DevelopmentKnowledge Fund awarded to M.R.S; U54GM104944 toJ.C.C., R01MH101054 to X.N.C., and R01MH109706 toE.C.O.; supported this work. The National Institutes of Gen-eral Medical Sciences (NIGMS) supports research infra-structure through Institutional Development Awards(IDeAs) and Center of Biomedical Research Excellence(COBRE) awards. This work stems from a collaborationof the Nevada Institute of Personalized Medicine (NIPM)and the Center for Neurodegeneration and TranslationalNeuroscience (CNTN). The collaboration is based on CO-BRE awards to the Cleveland Clinic Lou Ruvo Center forBrain Health and the University of Nevada Las Vegas(UNLV).

Uncited figure and references

Figs. 1 and 2; [298-302].

1551

1552

1553

1554

1555

1556

1557

1558

1559

1560

1561

RESEARCH IN CONTEXT

1. Systematic review: ---.

2. Interpretation: ---.

3. Future directions---.

1562

1563

1564

1565

1566

1567

1568

1569

1570

1571

1572

1573

1574

1575

1576

1577

1578

1579

1580

1581

1582

References

[1] Balla S, Thapar V, Verma S, Luong T, Faghri T, Huang CH, et al. Min-

imotif Miner: a tool for investigating protein function. Nat Methods

2006;3:175–7.

[2] Mi T, Merlin JC, Deverasetty S, Gryk MR, Bill TJ, Brooks AW, et al.

Minimotif Miner 3.0: database expansion and significantly improved

reduction of false-positive predictions from consensus sequences.

Nucleic Acids Res 2012;40:D252–60.

[3] Rajasekaran S, Balla S, Gradie P, Gryk MR, Kadaveru K, Kundeti V,

et al. Minimotif miner 2nd release: a database and web system for

motif search. Nucleic Acids Res 2009;37:D185–90.

[4] Lyon KF, Cai X, Young RJ, Mamun A-A, Rajasekaran S,

Schiller MR. Minimotif Miner 4: a million peptide minimotifs and

counting. Nucleic Acids Res 2018;46:D465–70.

FLA 5.5.0 DTD � TRCI168_proof

[5] Dinkel H, Van Roey K, Michael S, Kumar M, Uyar B, Altenberg B,

et al. ELM 2016-data update and new functionality of the eukaryotic

linear motif resource. Nucleic Acids Res 2016;44:D294–300.

[6] Tompa P, Davey NE, Gibson TJ, BabuMM. A million peptide motifs

for the molecular biologist. Mol Cell 2014;55:161–9.

[7] Lyon KF, Strong CL, Schooler SG, Young RJ, Roy N, Ozar B, et al.

Natural variability of minimotifs in 1092 people indicates that

minimotifs are targets of evolution. Nucleic Acids Res 2015;

43:6399–412. Q

[8] Reimand J, Wagih O, Bader GD. Evolutionary constraint and disease

associations of post-translational modification sites in human

genomes. PLoS Genet 2015;11:e1004919.

[9] Uyar B, Weatheritt RJ, Dinkel H, Davey NE, Gibson TJ. Proteome-

wide analysis of human disease mutations in short linear motifs:

neglected players in cancer? Mol Biosyst 2014;10:2626–42.

[10] Sobhy H. A review of functional motifs utilized by viruses.

Proteomes 2016;4. Q

[11] Kadaveru K, Vyas J, Schiller MR. Viral infection and human disease-

insights from minimotifs. Front Biosci 2008;13:6455–71.

[12] Choong NW, Cohen EEW. Forthcoming receptor tyrosine kinase

inhibitors. Expert Opin Ther Targets 2006;10:793–7.

[13] Przedborski S, Vila M, Jackson-Lewis V. Neurodegeneration: what is

it and where are we? J Clin Invest 2003;111:3–10.

[14] Montecchi-Palazzi L, Beavis R, Binz P-A, Chalkley RJ, Cottrell J,

Creasy D, et al. The PSI-MOD community standard for representa-

tion of protein modification data. Nat Biotechnol 2008;26:864–6.

[15] Schachter H. The joys of HexNAc. The synthesis and function of

N- and O-glycan branches. Glycoconj J 2000;17:465–83.

[16] Yan A, Lennarz WJ. Unraveling the mechanism of protein N-glyco-

sylation. J Biol Chem 2005;280:3121–4.

[17] Ohtsubo K, Marth JD. Glycosylation in cellular mechanisms of

health and disease. Cell 2006;126:855–67.

[18] Sol�a RJ, Griebenow K. Effects of glycosylation on the stability of

protein pharmaceuticals. J Pharm Sci 2009;98:1223–45.

[19] Grasbon-Frodl E, Lorenz H, Mann U, Nitsch RM, Windl O,

Kretzschmar HA. Loss of glycosylation associated with the T183A

mutation in human prion disease. Acta Neuropathol 2004;

108:476–84.

[20] Otvos L, Cudic M. Post-translational modifications in prion proteins.

Curr Protein Pept Sci 2002;3:643–52.

[21] Ermonval M, Mouillet-Richard S, Codogno P, Kellermann O, Botti J.

Evolving views in prion glycosylation: functional and pathological

implications. Biochimie 2003;85:33–45.

[22] DeArmond SJ, S�anchez H, Yehiely F, Qiu Y, Ninchak-Casey A,

Daggett V, et al. Selective neuronal targeting in prion disease. Neuron

1997;19:1337–48.

[23] Giuliano F, Bannwarth S, Monnot S, Cano A, Chabrol B, Vialettes B,

et al. Wolfram syndrome in French population: characterization of

novel mutations and polymorphisms in the WFS1 gene. Hum Mutat

2005;25:99–100.

[24] Meli M, Gasset M, Colombo G. Dynamic diagnosis of familial prion

diseases supports the b2-a2 loop as a Universal Interference Target.

PLoS ONE 2011;6:e19093.

[25] Cheng CJ, Daggett V. Different misfolding mechanisms converge on

common conformational changes: human prion protein pathogenic

mutants Y218N and E196K. Prion 2014;8:125–35.

[26] Gizaw ST, Koda T, AmanoM, KamimuraK, Ohashi T, HinouH, et al.

A comprehensive glycome profiling of Huntington’s disease trans-

genic mice. Biochim Biophys Acta 2015;1850:1704–18.

[27] Abou-Abbass H, Abou-El-Hassan H, BahmadH, Zibara K, Zebian A,

Youssef R, et al. Glycosylation and other PTMs alterations in neuro-

degenerative diseases: Current status and future role in neurotrauma.

Electrophoresis 2016;37:1549–61.

[28] Silveyra M-X, Cuadrado-Corrales N, Marcos A, Barquero M-S,

R�abano A, Calero M, et al. Altered glycosylation of acetylcholines-

terase in Creutzfeldt-Jakob disease. J Neurochem 2006;96:97–104.

� 25 July 2018 � 9:16 am � ce

1583

Page 13: Minimotifs dysfunction is pervasive in neurodegenerative

S. Sharma et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions - (2018) 1-19 13

1584

1585

1586

1587

1588

1589

1590

1591

1592

1593

1594

1595

1596

1597

1598

1599

1600

1601

1602

1603

1604

1605

1606

1607

1608

1609

1610

1611

1612

1613

1614

1615

1616

1617

1618

1619

1620

1621

1622

1623

1624

1625

1626

1627

1628

1629

1630

1631

1632

1633

1634

1635

1636

1637

1638

1639

1640

1641

1642

1643

1644

1645

1646

1647

1648

1649

1650

1651

1652

1653

1654

1655

1656

1657

1658

1659

1660

1661

1662

1663

1664

1665

1666

1667

1668

1669

1670

1671

1672

1673

1674

1675

1676

1677

1678

1679

1680

1681

1682

1683

1684

1685

1686

1687

1688

1689

1690

1691

1692

1693

1694

1695

1696

1697

1698

1699

1700

1701

1702

1703

1704

1705

1706

1707

1708

1709

1710

1711

1712

1713

1714

1715

1716

[29] Yan SD, Chen X, Schmidt AM, Brett J, Godman G, Zou YS, et al.

Glycated tau protein in Alzheimer disease: a mechanism for induc-

tion of oxidant stress. Proc Natl Acad Sci U S A 1994;91:7787–91.

[30] Liu F, Zaidi T, Iqbal K, Grundke-Iqbal I, Gong C-X. Aberrant glyco-

sylation modulates phosphorylation of tau by protein kinase A and

dephosphorylation of tau by protein phosphatase 2A and 5. Neurosci-

ence 2002;115:829–37.

[31] Sato Y, Naito Y, Grundke-Iqbal I, Iqbal K, Endo T. Analysis of N-gly-

cans of pathological tau: possible occurrence of aberrant processing

of tau in Alzheimer’s disease. FEBS Lett 2001;496:152–60.

[32] Hock E-M, Polymenidou M. Prion-like propagation as a pathogenic

principle in frontotemporal dementia. J Neurochem 2016;138 Suppl

1:163–83.

[33] Gong C-X, Iqbal K. Hyperphosphorylation of microtubule-

associated protein tau: a promising therapeutic target for Alzheimer

disease. Curr Med Chem 2008;15:2321–8.

[34] Wang J-Z, Xia Y-Y, Grundke-Iqbal I, Iqbal K. Abnormal hyperphos-

phorylation of tau: sites, regulation, and molecular mechanism of

neurofibrillary degeneration. J Alzheimers Dis 2013;33 Suppl

1:S123–39.

[35] Sj€ogren M, Davidsson P, Tullberg M, Minthon L, Wallin A,

Wikkelso C, et al. Both total and phosphorylated tau are increased

in Alzheimer’s disease. J Neurol Neurosurg Psychiatry 2001;

70:624–30.

[36] Hampel H, Teipel SJ. Total and phosphorylated tau proteins: evalua-

tion as core biomarker candidates in frontotemporal dementia. De-

ment Geriatr Cogn Disord 2004;17:350–4.

[37] Anderson JP, Walker DE, Goldstein JM, de Laat R, Banducci K,

Caccavello RJ, et al. Phosphorylation of Ser-129 is the dominant

pathological modification of alpha-synuclein in familial and sporadic

Lewy body disease. J Biol Chem 2006;281:29739–52.

[38] Tenreiro S, Eckermann K, Outeiro TF. Protein phosphorylation in

neurodegeneration: friend or foe? Front Mol Neurosci 2014;7:42.

[39] Fujiwara H, Hasegawa M, Dohmae N, Kawashima A, Masliah E,

Goldberg MS, et al. alpha-Synuclein is phosphorylated in synuclein-

opathy lesions. Nat Cell Biol 2002;4:160–4.

[40] Qing H, Wong W, McGeer EG, McGeer PL. Lrrk2 phosphorylates

alpha synuclein at serine 129: Parkinson disease implications. Bio-

chem Biophys Res Commun 2009;387:149–52.

[41] Inglis KJ, Chereau D, Brigham EF, Chiou S-S, Sch€obel S, Frigon NL,

et al. Polo-like kinase 2 (PLK2) phosphorylates alpha-synuclein at

serine 129 in central nervous system. J Biol Chem 2009;

284:2598–602.

[42] Okochi M,Walter J, Koyama A, Nakajo S, BabaM, Iwatsubo T, et al.

Constitutive phosphorylation of the Parkinson’s disease associated

alpha-synuclein. J Biol Chem 2000;275:390–7.

[43] Pronin AN, Morris AJ, Surguchov A, Benovic JL. Synucleins are a

novel class of substrates for G protein-coupled receptor kinases. J

Biol Chem 2000;275:26515–22.

[44] Kahle PJ, Neumann M, Ozmen L, Muller V, Jacobsen H,

Spooren W, et al. Hyperphosphorylation and insolubility of alpha-

synuclein in transgenic mouse oligodendrocytes. EMBO Rep

2002;3:583–8.

[45] Paleologou KE, Oueslati A, Shakked G, Rospigliosi CC, Kim H-Y,

Lamberto GR, et al. Phosphorylation at S87 is enhanced in synuclei-

nopathies, inhibits alpha-synuclein oligomerization, and influences

synuclein-membrane interactions. J Neurosci 2010;30:3184–98.

[46] Brooks J, Ding J, Simon-Sanchez J, Paisan-Ruiz C, Singleton AB,

Scholz SW. Parkin and PINK1 mutations in early-onset Parkinson’s

disease: comprehensive screening in publicly available cases and

control. J Med Genet 2009;46:375–81.

[47] Sha D, Chin L-S, Li L. Phosphorylation of parkin by Parkinson

disease-linked kinase PINK1 activates parkin E3 ligase function

and NF-kB signaling. Hum Mol Genet 2010;19:352–63.

[48] Kazlauskaite A, Kelly V, Johnson C, Baillie C, Hastie CJ, Peggie M,

et al. Phosphorylation of Parkin at Serine65 is essential for activation:

FLA 5.5.0 DTD � TRCI168_proof

elaboration of a Miro1 substrate-based assay of Parkin E3 ligase ac-

tivity. Open Biol 2014;4:130213.

[49] Dawson TM, Dawson VL. The role of parkin in familial and sporadic

Parkinson’s disease. Mov Disord 2010;25 Suppl 1:S32–9.

[50] Piccoli G, Onofri F, Cirnaru MD, Kaiser CJO, Jagtap P,

Kastenm€uller A, et al. Leucine-rich repeat kinase 2 binds to neuronal

vesicles through protein interactions mediated by its C-terminal

WD40 domain. Mol Cell Biol 2014;34:2147–61.

[51] Zoghbi HY, Orr HT. Pathogenic mechanisms of a polyglutamine-

mediated neurodegenerative disease, spinocerebellar ataxia type 1.

J Biol Chem 2009;284:7425–9.

[52] Chen H-K, Fernandez-Funez P, Acevedo SF, Lam YC, Kaytor MD,

Fernandez MH, et al. Interaction of Akt-phosphorylated ataxin-1

with 14-3-3 mediates neurodegeneration in spinocerebellar ataxia

type 1. Cell 2003;113:457–68.

[53] FayJM,ZhuC,ProctorEA,TaoY,CuiW,KeH,etal.Aphosphomimeticmu-

tationstabilizesSOD1andrescuescellviabilityinthecontextofanALS-

associatedmutation.Structure2016;24:1898–906.

[54] Resh MD. Targeting protein lipidation in disease. Trends Mol Med

2012;18:206–14.

[55] Lemonidis K, Sanchez-Perez MC, Chamberlain LH. Identification of

a novel sequence motif recognized by the ankyrin repeat domain of

zDHHC17/13 S-Acyltransferases. J Biol Chem 2015;290:21939–50.

[56] Tsutsumi R, Fukata Y, Fukata M. Discovery of protein-

palmitoylating enzymes. Pflugers Arch 2008;456:1199–206.

[57] Fukata Y, Fukata M. Protein palmitoylation in neuronal development

and synaptic plasticity. Nat Rev Neurosci 2010;11:161–75.

[58] Young FB, Butland SL, Sanders SS, Sutton LM, HaydenMR. Putting

proteins in their place: palmitoylation in Huntington disease and

other neuropsychiatric diseases. Prog Neurobiol 2012;97:220–38.

[59] Hornemann T. Palmitoylation and depalmitoylation defects. J Inherit

Metab Dis 2015;38:179–86.

[60] Fukata M, Fukata Y, Adesnik H, Nicoll RA, Bredt DS. Identification

of PSD-95 palmitoylating enzymes. Neuron 2004;44:987–96.

[61] Cho E, Park M. Palmitoylation in Alzheimer’s disease and other

neurodegenerative diseases. Pharmacol Res 2016;111:133–51.

[62] Martin DDO, Hayden MR. Post-translational myristoylation at the

cross roads of cell death, autophagy and neurodegeneration. Biochem

Soc Trans 2015;43:229–34.

[63] Valle C, Carr�ı MT. Cysteine modifications in the pathogenesis of

ALS. Front Mol Neurosci 2017;10:5.

[64] Antinone SE, Ghadge GD, Lam TT, Wang L, Roos RP, Green WN.

Palmitoylation of superoxide dismutase 1 (SOD1) is increased for fa-

milial amyotrophic lateral sclerosis-linked SOD1 mutants. J Biol

Chem 2013;288:21606–17.

[65] Choi J, Rees HD,Weintraub ST, Levey AI, Chin L-S, Li L. Oxidative

modifications and aggregation of Cu,Zn-superoxide dismutase asso-

ciated with Alzheimer and Parkinson diseases. J Biol Chem 2005;

280:11648–55.

[66] Greaves J, Lemonidis K, Gorleku OA, Cruchaga C, Grefen C,

Chamberlain LH. Palmitoylation-induced aggregation of cysteine-

string protein mutants that cause neuronal ceroid lipofuscinosis. J

Biol Chem 2012;287:37330–9.

[67] Fantini J, Yahi N. Molecular basis for the glycosphingolipid-binding

specificity of a-synuclein: key role of tyrosine 39 in membrane inser-

tion. J Mol Biol 2011;408:654–69.

[68] Fantini J, Carlus D, Yahi N. The fusogenic tilted peptide (67-78) of a-

synuclein is a cholesterol binding domain. Biochim Biophys Acta

2011;1808:2343–51.

[69] Okada T, Wakabayashi M, Ikeda K, Matsuzaki K. Formation of toxic

fibrils of Alzheimer’s amyloid beta-protein-(1-40) by monosialogan-

glioside GM1, a neuronal membrane component. J Mol Biol 2007;

371:481–9.

[70] Martinez Z, Zhu M, Han S, Fink AL. GM1 specifically interacts with

alpha-synuclein and inhibits fibrillation. Biochemistry 2007;

46:1868–77.

� 25 July 2018 � 9:16 am � ce

1717

Page 14: Minimotifs dysfunction is pervasive in neurodegenerative

S. Sharma et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions - (2018) 1-1914

1718

1719

1720

1721

1722

1723

1724

1725

1726

1727

1728

1729

1730

1731

1732

1733

1734

1735

1736

1737

1738

1739

1740

1741

1742

1743

1744

1745

1746

1747

1748

1749

1750

1751

1752

1753

1754

1755

1756

1757

1758

1759

1760

1761

1762

1763

1764

1765

1766

1767

1768

1769

1770

1771

1772

1773

1774

1775

1776

1777

1778

1779

1780

1781

1782

1783

1784

1785

1786

1787

1788

1789

1790

1791

1792

1793

1794

1795

1796

1797

1798

1799

1800

1801

1802

1803

1804

1805

1806

1807

1808

1809

1810

1811

1812

1813

1814

1815

1816

1817

1818

1819

1820

1821

1822

1823

1824

1825

1826

1827

1828

1829

1830

1831

1832

1833

1834

1835

1836

1837

1838

1839

1840

1841

1842

1843

1844

1845

1846

1847

1848

1849

1850

1851

[71] Jamin N, Neumann J-M, Ostuni MA, Vu TKN, Yao Z-X, Murail S,

et al. Characterization of the cholesterol recognition amino acid

consensus sequence of the peripheral-type benzodiazepine receptor.

Mol Endocrinol 2005;19:588–94.

[72] Hottman DA, Li L. Protein prenylation and synaptic plasticity: impli-

cations for Alzheimer’s disease. Mol Neurobiol 2014;50:177–85.

[73] MattsonMP. Methylation and acetylation in nervous system develop-

ment and neurodegenerative disorders. Ageing Res Rev 2003;

2:329–42.

[74] Drazic A, Myklebust LM, Ree R, Arnesen T. The world of protein

acetylation. Biochim Biophys Acta 2016;1864:1372–401.

[75] Thomas SN, Funk KE, Wan Y, Liao Z, Davies P, Kuret J, et al. Dual

modification of Alzheimer’s disease PHF-tau protein by lysine

methylation and ubiquitylation: a mass spectrometry approach.

Acta Neuropathol 2012;123:105–17.

[76] Min S-W, Cho S-H, Zhou Y, Schroeder S, Haroutunian V,

Seeley WW, et al. Acetylation of tau inhibits its degradation and con-

tributes to tauopathy. Neuron 2010;67:953–66.

[77] Min S-W, Chen X, Tracy TE, Li Y, Zhou Y, Wang C, et al. Critical

role of acetylation in tau-mediated neurodegeneration and cognitive

deficits. Nat Med 2015;21:1154–62.

[78] Trexler AJ, Rhoades E. N-Terminal acetylation is critical for forming

a-helical oligomer of a-synuclein. Protein Sci 2012;21:601–5.

[79] Bartels T, Kim NC, Luth ES, Selkoe DJ. N-alpha-acetylation of a-

synuclein increases its helical folding propensity, GM1 binding spec-

ificity and resistance to aggregation. PLoS One 2014;9:e103727.

[80] Arnesen T, Starheim KK, Van Damme P, Evjenth R, Dinh H,

Betts MJ, et al. The chaperone-like protein HYPK acts together

with NatA in cotranslational N-terminal acetylation and prevention

of Huntingtin aggregation. Mol Cell Biol 2010;30:1898–909.

[81] AsaumiM, IijimaK, Sumioka A, Iijima-AndoK, KirinoY, Nakaya T,

et al. Interaction of N-terminal acetyltransferasewith the cytoplasmic

domain of beta-amyloid precursor protein and its effect on A beta

secretion. J Biochem 2005;137:147–55.

[82] Holmes WM, Mannakee BK, Gutenkunst RN, Serio TR. Loss of

amino-terminal acetylation suppresses a prion phenotype by modu-

lating global protein folding. Nat Commun 2014;5:4383.

[83] Pritzker LB, Joshi S, Harauz G, Moscarello MA. Deimination of

myelin basic protein. 2. Effect of methylation of MBP on its deimina-

tion by peptidylarginine deiminase. Biochemistry 2000;39:5382–8.

[84] Gu L, Robinson RAS. Proteomic approaches to quantify cysteine

reversible modifications in aging and neurodegenerative diseases.

Proteomics Clin Appl 2016;10:1159–77.

[85] Dr€ogeW. Oxidative stress and ageing: is ageing a cysteine deficiency

syndrome? Philos Trans R Soc Lond B Biol Sci 2005;360:2355–72.

[86] Li J, OW, LiW, Jiang Z-G, Ghanbari HA. Oxidative stress and neuro-

degenerative disorders. Int J Mol Sci 2013;14:24438–75.

[87] Barber SC, Shaw PJ. Oxidative stress in ALS: key role in motor

neuron injury and therapeutic target. Free Radic Biol Med 2010;

48:629–41.

[88] Furukawa Y, O’Halloran TV. Posttranslational modifications in

Cu,Zn-superoxide dismutase and mutations associated with amyotro-

phic lateral sclerosis. Antioxid Redox Signal 2006;8:847–67.

[89] McAlary L, Yerbury JJ, Aquilina JA. Glutathionylation potentiates

benign superoxide dismutase 1 variants to the toxic forms associated

with amyotrophic lateral sclerosis. Sci Rep 2013;3:3275.

[90] Wilcox KC, Zhou L, Jordon JK, Huang Y, Yu Y, Redler RL, et al.

Modifications of superoxide dismutase (SOD1) in human erythro-

cytes: a possible role in amyotrophic lateral sclerosis. J Biol Chem

2009;284:13940–7.

[91] Nakamura T, Tu S, AkhtarMW, Sunico CR, Okamoto S-I, Lipton SA.

Aberrant protein s-nitrosylation in neurodegenerative diseases.

Neuron 2013;78:596–614.

[92] Smith CD, Carney JM, Starke-Reed PE, Oliver CN, Stadtman ER,

Floyd RA, et al. Excess brain protein oxidation and enzyme dysfunc-

tion in normal aging and in Alzheimer disease. Proc Natl Acad Sci U

S A 1991;88:10540–3.

FLA 5.5.0 DTD � TRCI168_proof

[93] Danielson SR, Andersen JK. Oxidative and nitrative protein modifi-

cations in Parkinson’s disease. Free Radic Biol Med 2008;

44:1787–94.

[94] Giasson BI, Ischiropoulos H, Lee VM-Y, Trojanowski JQ. The rela-

tionship between oxidative/nitrative stress and pathological inclu-

sions in Alzheimer’s and Parkinson’s diseases. Free Radic Biol

Med 2002;32:1264–75.

[95] Stamler JS, Toone EJ, Lipton SA, Sucher NJ. (S)NO signals: translo-

cation, regulation, and a consensus motif. Neuron 1997;18:691–6.

[96] Stamler JS. Redox signaling: nitrosylation and related target interac-

tions of nitric oxide. Cell 1994;78:931–6.

[97] Nakamura T, Lipton SA. S-Nitrosylation and uncompetitive/fast off-

rate (UFO) drug therapy in neurodegenerative disorders of protein

misfolding. Cell Death Differ 2007;14:1305–14.

[98] Dantuma NP, Bott LC. The ubiquitin-proteasome system in neurode-

generative diseases: precipitating factor, yet part of the solution.

Front Mol Neurosci 2014;7:70.

[99] Shimura H, Schlossmacher MG, Hattori N, Frosch MP,

Trockenbacher A, Schneider R, et al. Ubiquitination of a new form

of alpha-synuclein by parkin from human brain: implications for Par-

kinson’s disease. Science 2001;293:263–9.

[100] Chung KK, Zhang Y, Lim KL, Tanaka Y, Huang H, Gao J, et al. Par-

kin ubiquitinates the alpha-synuclein-interacting protein, synphilin-

1: implications for Lewy-body formation in Parkinson disease. Nat

Med 2001;7:1144–50.

[101] Wang GP, Khatoon S, Iqbal K, Grundke-Iqbal I. Brain ubiquitin is

markedly elevated in Alzheimer disease. Brain Res 1991;

566:146–51.

[102] Kurita K, Manaka H, Kato T, Katagiri T, Sasaki H. [A case of

Creutzfeldt-Jakob diseasewith markedly elevated ubiquitin concentra-

tion in the cerebrospinal fluid]. Rinsho Shinkeigaku 1991;31:666–8.

[103] Kang S, Hong S. Molecular pathogenesis of spinocerebellar ataxia

type 1 disease. Mol Cells 2009;27:621–7.

[104] Shibata H, Huynh DP, Pulst SM. A novel protein with RNA-binding

motifs interacts with ataxin-2. Hum Mol Genet 2000;9:1303–13.

[105] McFadden K, Hamilton RL, Insalaco SJ, Lavine L, Al-Mateen M,

WangG, et al. Neuronal intranuclear inclusion diseasewithout polyglut-

amine inclusions in a child. J Neuropathol ExpNeurol 2005;64:545–52.

[106] Nath SR, Lieberman AP. The ubiquitination, disaggregation and pro-

teasomal degradation machineries in polyglutamine disease. Front

Mol Neurosci 2017;10:78.

[107] Sambataro F, Pennuto M. Post-translational modifications and pro-

tein quality control in motor neuron and polyglutamine diseases.

Front Mol Neurosci 2017;10:82.

[108] Gray KM, Kaifer KA, Baillat D,Wen Y, Bonacci TR, Ebert AD, et al.

Self-oligomerization regulates stability of Survival Motor Neuron

(SMN) protein isoforms by sequestering an SCFSlmb degron. Mol

Biol Cell 2018;29:96–110.

[109] Burnett BG, Mu~noz E, Tandon A, Kwon DY, Sumner CJ,

Fischbeck KH. Regulation of SMN protein stability. Mol Cell Biol

2009;29:1107–15.

[110] Yang Y, He Y, Wang X, Liang Z, He G, Zhang P, et al. Protein SU-

MOylation modification and its associations with disease. Open

Biol 2017;7.

[111] Anderson DB, Zanella CA, Henley JM, Cimarosti H. Sumoylation:

implications for neurodegenerative diseases. Adv Exp Med Biol

2017;963:261–81.

[112] Dorval V, Fraser PE. SUMO on the road to neurodegeneration. Bio-

chim Biophys Acta 2007;1773:694–706.

[113] Hoppe JB, Salbego CG, Cimarosti H. SUMOylation: novel neuropro-

tective approach for Alzheimer’s disease? Aging Dis 2015;6:322–30.

[114] Lee L, Sakurai M,Matsuzaki S, Arancio O, Fraser P. SUMO and Alz-

heimer’s disease. Neuromolecular Med 2013;15:720–36.

[115] Luo H-B, Xia Y-Y, Shu X-J, Liu Z-C, Feng Y, Liu X-H, et al. SU-

MOylation at K340 inhibits tau degradation through deregulating

its phosphorylation and ubiquitination. Proc Natl Acad Sci U S A

2014;111:16586–91.

� 25 July 2018 � 9:16 am � ce

Page 15: Minimotifs dysfunction is pervasive in neurodegenerative

S. Sharma et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions - (2018) 1-19 15

1852

1853

1854

1855

1856

1857

1858

1859

1860

1861

1862

1863

1864

1865

1866

1867

1868

1869

1870

1871

1872

1873

1874

1875

1876

1877

1878

1879

1880

1881

1882

1883

1884

1885

1886

1887

1888

1889

1890

1891

1892

1893

1894

1895

1896

1897

1898

1899

1900

1901

1902

1903

1904

1905

1906

1907

1908

1909

1910

1911

1912

1913

1914

1915

1916

1917

1918

1919

1920

1921

1922

1923

1924

1925

1926

1927

1928

1929

1930

1931

1932

1933

1934

1935

1936

1937

1938

1939

1940

1941

1942

1943

1944

1945

1946

1947

1948

1949

1950

1951

1952

1953

1954

1955

1956

1957

1958

1959

1960

1961

1962

1963

1964

1965

1966

1967

1968

1969

1970

1971

1972

1973

1974

1975

1976

1977

1978

1979

1980

1981

1982

1983

1984

1985

[116] Kontaxi C, Piccardo P, Gill AC. Lysine-directed post-translational

modifications of tau protein in Alzheimer’s disease and related tauo-

pathies. Front Mol Biosci 2017;4:56.

[117] Krumova P, Meulmeester E, Garrido M, Tirard M, Hsiao H-H,

Bossis G, et al. Sumoylation inhibits a-synuclein aggregation and

toxicity. J Cell Biol 2011;194:49–60.

[118] Kunadt M, Eckermann K, Stuendl A, Gong J, Russo B, Strauss K,

et al. Extracellular vesicle sorting of a-Synuclein is regulated by su-

moylation. Acta Neuropathol 2015;129:695–713.

[119] Steffan JS, Agrawal N, Pallos J, Rockabrand E, Trotman LC,

Slepko N, et al. SUMO modification of Huntingtin and Huntington’s

disease pathology. Science 2004;304:100–4.

[120] Dangoumau A, Marouillat S, Burlaud Gaillard J, Uzbekov R, Veyrat-

Durebex C, Blasco H, et al. Inhibition of pathogenic mutant SOD1

aggregation in cultured motor neuronal cells by prevention of its SU-

MOylation on Lysine 75. Neurodegener Dis 2016;16:161–71.

[121] Dangoumau A, Veyrat-Durebex C, Blasco H, Praline J, Corcia P,

Andres CR, et al. Protein SUMOylation, an emerging pathway in

amyotrophic lateral sclerosis. Int J Neurosci 2013;123:366–74.

[122] Niikura T, Kita Y, Abe Y. SUMO3 modification accelerates the

aggregation of ALS-linked SOD1 mutants. PLoS One 2014;

9:e101080.

[123] Wang T, Xu W, Qin M, Yang Y, Bao P, Shen F, et al. Pathogenic mu-

tations in the valosin-containing protein/p97(VCP) N-domain Inhibit

the SUMOylation of VCP and lead to impaired stress response. J Biol

Chem 2016;291:14373–84.

[124] MoscarelloMA,Mastronardi FG,Wood DD. The role of citrullinated

proteins suggests a novel mechanism in the pathogenesis of multiple

sclerosis. Neurochem Res 2007;32:251–6.

[125] Jang B, Ishigami A, Maruyama N, Carp RI, Kim Y-S, Choi E-K. Pep-

tidylarginine deiminase and protein citrullination in prion diseases:

strong evidence of neurodegeneration. Prion 2013;7:42–6.

[126] Ishigami A, Ohsawa T, Hiratsuka M, Taguchi H, Kobayashi S,

Saito Y, et al. Abnormal accumulation of citrullinated proteins

catalyzed by peptidylarginine deiminase in hippocampal extracts

from patients with Alzheimer’s disease. J Neurosci Res 2005;

80:120–8.

[127] Nicholas AP. Dual immunofluorescence study of citrullinated pro-

teins in Parkinson diseased substantia nigra. Neurosci Lett 2011;

495:26–9.

[128] Jang B, Jeon Y-C, Choi J-K, Park M, Kim J-I, Ishigami A, et al. Pep-

tidylarginine deiminase modulates the physiological roles of enolase

via citrullination: links between altered multifunction of enolase and

neurodegenerative diseases. Biochem J 2012;445:183–92.

[129] Acharya NK, Nagele EP, Han M, Coretti NJ, DeMarshall C,

Kosciuk MC, et al. Neuronal PAD4 expression and protein citrullina-

tion: possible role in production of autoantibodies associated with

neurodegenerative disease. J Autoimmun 2012;38:369–80.

[130] Yang L, Tan D, Piao H.Myelin basic protein citrullination in multiple

sclerosis: a potential therapeutic target for the pathology. Neurochem

Res 2016;41:1845–56.

[131] Kim JK, Mastronardi FG, Wood DD, Lubman DM, Zand R,

Moscarello MA. Multiple sclerosis: an important role for post-

translational modifications of myelin basic protein in pathogenesis.

Mol Cell Proteomics 2003;2:453–62.

[132] Ciechanover A, Kwon YT. Degradation of misfolded proteins in

neurodegenerative diseases: therapeutic targets and strategies. Exp

Mol Med 2015;47:e147.

[133] Wilson CM, Mushtaq G, Kamal MA, Terro F. The Role of endopro-

teolytic processing in neurodegeneration. CNS Neurol Disord Drug

Targets 2016;15:1222–30.

[134] Marcelli S, CorboM, Iannuzzi F, Negri L, Blandini F, Nistic�o R, et al.

The involvement of post-translational modifications in Alzheimer’s

disease. Curr Alzheimer Res 2018;15:313–35.

[135] Wang X, Zhou X, Li G, Zhang Y, Wu Y, Song W. Modifications and

trafficking of APP in the pathogenesis of Alzheimer’s disease. Front

Mol Neurosci 2017;10:294.

FLA 5.5.0 DTD � TRCI168_proof

[136] Vidal R, Frangione B, Rostagno A, Mead S, R�ev�esz T, Plant G, et al.

A stop-codon mutation in the BRI gene associated with familial

British dementia. Nature 1999;399:776–81.

[137] Kim SH, Wang R, Gordon DJ, Bass J, Steiner DF, Lynn DG, et al.

Furin mediates enhanced production of fibrillogenic ABri peptides

in familial British dementia. Nat Neurosci 1999;2:984–8.

[138] Kim S-H, Creemers JWM, Chu S, Thinakaran G, Sisodia SS. Proteo-

lytic processing of familial British dementia-associated BRI variants:

evidence for enhanced intracellular accumulation of amyloidogenic

peptides. J Biol Chem 2002;277:1872–7.

[139] Kim SH,Wang R, GordonDJ, Bass J, Steiner DF, ThinakaranG, et al.

Familial British dementia: expression and metabolism of BRI. Ann N

YAcad Sci 2000;920:93–9.

[140] Kim YJ, Yi Y, Sapp E, Wang Y, Cuiffo B, Kegel KB, et al. Caspase 3-

cleaved N-terminal fragments of wild-type and mutant huntingtin are

present in normal and Huntington’s disease brains, associate with

membranes, and undergo calpain-dependent proteolysis. Proc Natl

Acad Sci U S A 2001;98:12784–9.

[141] Sun B, Fan W, Balciunas A, Cooper JK, Bitan G, Steavenson S, et al.

Polyglutamine repeat length-dependent proteolysis of huntingtin.

Neurobiol Dis 2002;11:111–22.

[142] Landles C, Sathasivam K, Weiss A, Woodman B, Moffitt H,

Finkbeiner S, et al. Proteolysis of mutant huntingtin produces an

exon 1 fragment that accumulates as an aggregated protein in

neuronal nuclei in Huntington disease. J Biol Chem 2010;

285:8808–23.

[143] Miller JP, Holcomb J, Al-Ramahi I, de Haro M, Gafni J, Zhang N,

et al. Matrix metalloproteinases are modifiers of huntingtin proteoly-

sis and toxicity in Huntington’s disease. Neuron 2010;67:199–212.

[144] Ratovitski T, Chighladze E, Waldron E, Hirschhorn RR, Ross CA.

Cysteine proteases bleomycin hydrolase and cathepsin Z mediate

N-terminal proteolysis and toxicity of mutant huntingtin. J Biol

Chem 2011;286:12578–89.

[145] Wang X, Huang T, Bu G, Xu H. Dysregulation of protein trafficking

in neurodegeneration. Mol Neurodegener 2014;9:31.

[146] Sharma S, Toledo O, Hedden M, Lyon KF, Brooks SB, David RP,

et al. The Functional Human C-Terminome. PLoS One 2016;

11:e0152731.

[147] Schreij AMA, Fon EA, McPherson PS. Endocytic membrane traf-

ficking and neurodegenerative disease. Cell Mol Life Sci 2016;

73:1529–45.

[148] Abeliovich A, Gitler AD. Defects in trafficking bridge Parkinson’s

disease pathology and genetics. Nature 2016;539:207–16.

[149] Hasegawa T, Sugeno N, Kikuchi A, Baba T, Aoki M. Membrane traf-

ficking illuminates a path to Parkinson’s disease. Tohoku J Exp Med

2017;242:63–76.

[150] Esposito G, Ana Clara F, Verstreken P. Synaptic vesicle trafficking

and Parkinson’s disease. Dev Neurobiol 2012;72:134–44.

[151] Hunn BHM, Cragg SJ, Bolam JP, Spillantini M-G, Wade-Martins R.

Impaired intracellular trafficking defines early Parkinson’s disease.

Trends Neurosci 2015;38:178–88.

[152] Do CB, Tung JY, Dorfman E, Kiefer AK, Drabant EM, Francke U,

et al. Web-based genome-wide association study identifies two novel

loci and a substantial genetic component for Parkinson’s disease.

PLoS Genet 2011;7:e1002141.

[153] Martin I, Kim JW, Dawson VL, Dawson TM. LRRK2 pathobiology

in Parkinson’s disease. J Neurochem 2014;131:554–65.

[154] Matta S, Van Kolen K, da Cunha R, van den Bogaart G,

Mandemakers W, Miskiewicz K, et al. LRRK2 controls an EndoA

phosphorylation cycle in synaptic endocytosis. Neuron 2012;

75:1008–21.

[155] Park J-S, Kim D-H, Yoon S-Y. Regulation of amyloid precursor pro-

tein processing by its KFERQ motif. BMB Rep 2016;49:337–42.

[156] Wang J, Shan C, Cao W, Zhang C, Teng J, Chen J. SCG10 promotes

non-amyloidogenic processing of amyloid precursor protein by facil-

itating its trafficking to the cell surface. Hum Mol Genet 2013;

22:4888–900.

� 25 July 2018 � 9:16 am � ce

Page 16: Minimotifs dysfunction is pervasive in neurodegenerative

Q22

S. Sharma et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions - (2018) 1-1916

1986

1987

1988

1989

1990

1991

1992

1993

1994

1995

1996

1997

1998

1999

2000

2001

2002

2003

2004

2005

2006

2007

2008

2009

2010

2011

2012

2013

2014

2015

2016

2017

2018

2019

2020

2021

2022

2023

2024

2025

2026

2027

2028

2029

2030

2031

2032

2033

2034

2035

2036

2037

2038

2039

2040

2041

2042

2043

2044

2045

2046

2047

2048

2049

2050

2051

2052

2053

2054

2055

2056

2057

2058

2059

2060

2061

2062

2063

2064

2065

2066

2067

2068

2069

2070

2071

2072

2073

2074

2075

2076

2077

2078

2079

2080

2081

2082

2083

2084

2085

2086

2087

2088

2089

2090

2091

2092

2093

2094

2095

2096

2097

2098

2099

2100

2101

2102

2103

2104

2105

2106

2107

2108

2109

2110

2111

2112

2113

2114

2115

2116

2117

2118

[157] Munro S, Pelham HRB. A C-terminal signal prevents secretion of

luminal ER proteins. Cell 1987;48:899–907.

[158] Wang P, Li B, Zhou L, Fei E, Wang G. The KDEL receptor induces

autophagy to promote the clearance of neurodegenerative disease-

related proteins. Neuroscience 2011;190:43–55.

[159] Lee J, Retamal C, Cuiti~no L, Caruano-Yzermans A, Shin J-E, van

Kerkhof P, et al. Adaptor protein sorting nexin 17 regulates amyloid

precursor protein trafficking and processing in the early endosomes. J

Biol Chem 2008;283:11501–8.

[160] van Kerkhof P, Lee J, McCormick L, Tetrault E, LuW, SchoenfishM,

et al. Sorting nexin 17 facilitates LRP recycling in the early endo-

some. EMBO J 2005;24:2851–61.

[161] Paulson HL. The spinocerebellar ataxias. J Neuroophthalmol 2009;

29:227–37.

[162] Deretic D, Williams AH, Ransom N, Morel V, Hargrave PA,

Arendt A. Rhodopsin C terminus, the site of mutations causing retinal

disease, regulates trafficking by binding to ADP-ribosylation factor 4

(ARF4). Proc Natl Acad Sci USA 2005;102:3301–6.

[163] Deretic D, Schmerl S, Hargrave PA, Arendt A, McDowell JH. Regu-

lation of sorting and post-Golgi trafficking of rhodopsin by its C-ter-

minal sequence QVS(A)PA. Proc Natl Acad Sci U S A 1998;

95:10620–5.

[164] Ghavami S, Shojaei S, Yeganeh B, Ande SR, Jangamreddy JR,

Mehrpour M, et al. Autophagy and apoptosis dysfunction in neurode-

generative disorders. Prog Neurobiol 2014;112:24–49.

[165] Cuervo AM, Stefanis L, Fredenburg R, Lansbury PT, Sulzer D.

Impaired degradation of mutant alpha-synuclein by chaperone-

mediated autophagy. Science 2004;305:1292–5.

[166] Alvarez-Erviti L, Seow Y, Schapira AHV, Rodriguez-Oroz MC,

Obeso JA, Cooper JM. Influence of microRNA deregulation on

chaperone-mediated autophagy and a-synuclein pathology in Parkin-

son’s disease. Cell Death Dis 2013;4:e545.

[167] Wang Y, Martinez-Vicente M, Kr€uger U, Kaushik S, Wong E,

Mandelkow E-M, et al. Tau fragmentation, aggregation and clear-

ance: the dual role of lysosomal processing. Hum Mol Genet 2009;

18:4153–70.

[168] . Witt S, Uversky V, eds. Protein chaperones and protection from

neurodegenerative diseases. Hoboken. John Wiley & Sons; 2011.

[169] Qi L, Zhang X-D, Wu J-C, Lin F, Wang J, DiFiglia M, et al. The role

of chaperone-mediated autophagy in huntingtin degradation. PLoS

One 2012;7:e46834.

[170] Kalay E, de Brouwer APM, Caylan R, Nabuurs SB, Wollnik B,

Karaguzel A, et al. A novel D458V mutation in the SANS PDZ bind-

ing motif causes atypical Usher syndrome. J Mol Med 2005;

83:1025–32.

[171] Yan J, Pan L, ChenX,Wu L, ZhangM. The structure of the harmonin/

sans complex reveals an unexpected interaction mode of the two

Usher syndrome proteins. Proc Natl Acad Sci U S A 2010;

107:4040–5.

[172] Lee G, Thangavel R, SharmaVM, Litersky JM, Bhaskar K, Fang SM,

et al. Phosphorylation of tau by fyn: implications for Alzheimer’s dis-

ease. J Neurosci 2004;24:2304–12.

[173] Lee G, Newman ST, Gard DL, Band H, Panchamoorthy G. Tau inter-

acts with src-family non-receptor tyrosine kinases. J Cell Sci 1998;

111:3167–77.

[174] Lee G. Tau and src family tyrosine kinases. Biochim Biophys Acta

2005;1739:323–30.

[175] Leal A, Huehne K, Bauer F, Sticht H, Berger P, Suter U, et al. Iden-

tification of the variant Ala335Val of MED25 as responsible for

CMT2B2: molecular data, functional studies of the SH3 recognition

motif and correlation between wild-type MED25 and PMP22 RNA

levels in CMT1A animal models. Neurogenetics 2009;10:275–87.

[176] Borg JP, Yang Y, De Tadd�eo-Borg M, Margolis B, Turner RS. The

X11alpha protein slows cellular amyloid precursor protein process-

ing and reduces Abeta40 and Abeta42 secretion. J Biol Chem

1998;273:14761–6.

FLA 5.5.0 DTD � TRCI168_proof

[177] King GD, Perez RG, Steinhilb ML, Gaut JR, Turner RS. X11alpha

modulates secretory and endocytic trafficking and metabolism of am-

yloid precursor protein: mutational analysis of the YENPTY

sequence. Neuroscience 2003;120:143–54.

[178] Ho A, Liu X, S€udhof TC. Deletion of Mint proteins decreases amy-

loid production in transgenic mouse models of Alzheimer’s disease. J

Neurosci 2008;28:14392–400.

[179] Chen H-K, Fernandez-Funez P, Acevedo SF, Lam YC, Kaytor MD,

Fernandez MH, et al. Interaction of Akt-phosphorylated ataxin-1

with 14-3-3 mediates neurodegeneration in spinocerebellar ataxia

type 1. Cell 2003;113:457–68.

[180] Barrett PJ, Song Y, Van Horn WD, Hustedt EJ, Schafer JM,

Hadziselimovic A, et al. The amyloid precursor protein has a flexible

transmembrane domain and binds cholesterol. Science 2012;

336:1168–71.

[181] Song Y, Hustedt EJ, Brandon S, Sanders CR. Competition between

homodimerization and cholesterol binding to the C99 domain of

the amyloid precursor protein. Biochemistry 2013;52:5051–64.

[182] Kim S, Jeon T-J, Oberai A, Yang D, Schmidt JJ, Bowie JU.

Transmembrane glycine zippers: physiological and pathological

roles in membrane proteins. Proc Natl Acad Sci U S A 2005;

102:14278–83.

[183] Knafo S, S�anchez-Puelles C, Palomer E, Delgado I, Draffin JE,

Mingo J, et al. PTEN recruitment controls synaptic and cognitive

function in Alzheimer’s models. Nat Neurosci 2016;19:443–53.

[184] Liu F, Iqbal K, Grundke-Iqbal I, Hart GW, Gong C-X. O-GlcNAcy-

lation regulates phosphorylation of tau: a mechanism involved in

Alzheimer’s disease. Proc Natl Acad Sci U S A 2004;101:10804–9.

[185] Dickey CA, Koren J, Zhang Y-J, Xu Y-F, Jinwal UK, Birnbaum MJ,

et al. Akt and CHIP coregulate tau degradation through coordinated

interactions. Proc Natl Acad Sci U S A 2008;105:3622–7.

[186] Ross CA, Poirier MA. Protein aggregation and neurodegenerative

disease. Nat Med 2004;10 Suppl:S10–7.

[187] Ben Yehuda A, Risheq M, Novoplansky O, Bersuker K, Kopito RR,

Goldberg M, et al. Ubiquitin accumulation on disease associated pro-

tein aggregates is correlated with nuclear ubiquitin depletion, histone

de-ubiquitination and impaired DNA damage response. PLoS One

2017;12:e0169054.

[188] Zhang Y-Q, Sarge KD. Sumoylation of amyloid precursor protein

negatively regulates Abeta aggregate levels. Biochem Biophys Res

Commun 2008;374:673–8.

[189] Rezaei-Ghaleh N, Amininasab M, Kumar S, Walter J,

Zweckstetter M. Phosphorylation modifies the molecular stability

of b-amyloid deposits. Nat Commun 2016;7:11359.

[190] �Simi�c G, Babi�c Leko M, Wray S, Harrington C, Delalle I, Jovanov-

Milo�sevi�c N, et al. Tau Protein hyperphosphorylation and aggrega-

tion in Alzheimer’s disease and other tauopathies, and possible neu-

roprotective strategies. Biomolecules 2016;6:6.

[191] Gorsky MK, Burnouf S, Dols J, Mandelkow E, Partridge L. Acetyla-

tion mimic of lysine 280 exacerbates human Tau neurotoxicity

in vivo. Sci Rep 2016;6:22685.

[192] Barrett PJ, Timothy Greenamyre J. Post-translational modification of

a-synuclein in Parkinson’s disease. Brain Res 2015;1628:247–53.

[193] Duce JA, Wong BX, Durham H, Devedjian J-C, Smith DP, Devos D.

Post translational changes to a-synuclein control iron and dopamine

trafficking; a concept for neuron vulnerability in Parkinson’s disease.

Mol Neurodegener 2017;12:45.

[194] Silva A, de Almeida AV, Macedo-Ribeiro S. Polyglutamine expan-

sion diseases: More than simple repeats. J Struct Biol 2018;

201:139–54.

[195] Saudou F, Humbert S. The biology of huntingtin. Neuron 2016;

89:910–26.

[196] Sargeant DP, Deverasetty S, Strong CL, Alaniz IJ, Bartlett A,

Brandon NR, et al. The HIVToolbox 2 web system integrates

sequence, structure, function and mutation analysis. PLoS One

2014;9:e98810.

� 25 July 2018 � 9:16 am � ce

2119

Page 17: Minimotifs dysfunction is pervasive in neurodegenerative

S. Sharma et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions - (2018) 1-19 17

2120

2121

2122

2123

2124

2125

2126

2127

2128

2129

2130

2131

2132

2133

2134

2135

2136

2137

2138

2139

2140

2141

2142

2143

2144

2145

2146

2147

2148

2149

2150

2151

2152

2153

2154

2155

2156

2157

2158

2159

2160

2161

2162

2163

2164

2165

2166

2167

2168

2169

2170

2171

2172

2173

2174

2175

2176

2177

2178

2179

2180

2181

2182

2183

2184

2185

2186

2187

2188

2189

2190

2191

2192

2193

2194

2195

2196

2197

2198

2199

2200

2201

2202

2203

2204

2205

2206

2207

2208

2209

2210

2211

2212

2213

2214

2215

2216

2217

2218

2219

2220

2221

2222

2223

2224

2225

2226

2227

2228

2229

2230

2231

2232

2233

2234

2235

2236

2237

2238

2239

2240

2241

2242

2243

2244

2245

2246

2247

2248

2249

2250

2251

2252

[197] Sarmady M, Dampier W, Tozeren A. Sequence- and interactome-

based prediction of viral protein hotspots targeting host proteins: a

case study for HIV Nef. PLoS One 2011;6:e20735.

[198] Xue B, Brown CJ, Dunker AK, Uversky VN. Intrinsically disordered

regions of p53 family are highly diversified in evolution. Biochim

Biophys Acta 2013;1834:725–38.

[199] Fernandez-Fernandez MR, Sot B. The relevance of protein-protein

interactions for p53 function: the CPE contribution. Protein Eng

Des Sel 2011;24:41–51.

[200] Visscher PM,WrayNR, ZhangQ, Sklar P,McCarthyMI, BrownMA,

et al. 10 Years of GWAS discovery: biology, function, and translation.

Am J Hum Genet 2017;101:5–22.

[201] Singleton A, Hardy J. The evolution of genetics: Alzheimer’s and

Parkinson’s diseases. Neuron 2016;90:1154–63.

[202] Guerreiro RJ, Gustafson DR, Hardy J. The genetic architecture of

Alzheimer’s disease: beyond APP, PSENs and APOE. Neurobiol Ag-

ing 2012;33:437–56.

[203] Gatz M, Pedersen NL, Berg S, Johansson B, Johansson K,

Mortimer JA, et al. Heritability for Alzheimer’s disease: the study

of dementia in Swedish twins. J Gerontol A Biol Sci Med Sci

1997;52:M117–25.

[204] Nicolas G, Charbonnier C, Campion D. From common to rare vari-

ants: the genetic component of Alzheimer disease. Hum Hered

2016;81:129–41.

[205] Rovelet-LecruxA, HannequinD, Raux G, LeMeur N, Laquerri�ere A,

Vital A, et al. APP locus duplication causes autosomal dominant

early-onset Alzheimer disease with cerebral amyloid angiopathy.

Nat Genet 2006;38:24–6.

[206] Sleegers K, Brouwers N, Gijselinck I, Theuns J, Goossens D,

Wauters J, et al. APP duplication is sufficient to cause early onset

Alzheimer’s dementia with cerebral amyloid angiopathy. Brain

2006;129:2977–83.

[207] Deming Y, Li Z, Kapoor M, Harari O, Del-Aguila JL, Black K, et al.

Genome-wide association study identifies four novel loci associated

with Alzheimer’s endophenotypes and disease modifiers. Acta Neu-

ropathol 2017;133:839–56.

[208] Guo Q, Wang Z, Li H, Wiese M, Zheng H. APP physiological and

pathophysiological functions: insights from animal models. Cell

Res 2012;22:78–89.

[209] Corti O, Lesage S, Brice A. What genetics tells us about the causes

and mechanisms of Parkinson’s disease. Physiol Rev 2011;

91:1161–218.

[210] Verstraeten A, Theuns J, Van Broeckhoven C. Progress in unraveling

the genetic etiology of Parkinson disease in a genomic era. Trends

Genet 2015;31:140–9.

[211] Nussbaum RL, Ellis CE. Alzheimer’s disease and Parkinson’s dis-

ease. N Engl J Med 2003;348:1356–64.

[212] Hardy J. Genetic analysis of pathways to Parkinson disease. Neuron

2010;68:201–6.

[213] Chang D, Nalls MA, Hallgr�ımsd�ottir IB, Hunkapiller J, van der

Brug M, Cai F, et al. A meta-analysis of genome-wide association

studies identifies 17 new Parkinson’s disease risk loci. Nat Genet

2017;49:1511–6.

[214] Doggett EA, Zhao J,Mork CN, HuD, Nichols RJ. Phosphorylation of

LRRK2 serines 955 and 973 is disrupted by Parkinson’s disease mu-

tations and LRRK2 pharmacological inhibition. J Neurochem 2012;

120:37–45.

[215] Javed H, KamalMA, Ojha S. An overview on the role of a -synuclein

in experimental models of Parkinson’s disease from pathogenesis to

therapeutics. CNS Neurol Disord Drug Targets 2016;15:1240–52.

[216] Tremblay M-�E, Lowery RL, Majewska AK. Microglial interactions

with synapses are modulated by visual experience. PLoS Biol

2010;8:e1000527.

[217] Wu Y, Dissing-Olesen L, MacVicar BA, Stevens B. Microglia: dy-

namic mediators of synapse development and plasticity. Trends Im-

munol 2015;36:605–13.

FLA 5.5.0 DTD � TRCI168_proof

[218] Lambert J-C, Heath S, Even G, Campion D, Sleegers K, Hiltunen M,

et al. Genome-wide association study identifies variants at CLU and

CR1 associated with Alzheimer’s disease. Nat Genet 2009;

41:1094–9.

[219] Lambert JC, Ibrahim-Verbaas CA, Harold D, Naj AC, Sims R,

Bellenguez C, et al. Meta-analysis of 74,046 individuals identifies

11 new susceptibility loci for Alzheimer’s disease. Nat Genet 2013;

45:1452–8.

[220] Hollingworth P, Harold D, Sims R, Gerrish A, Lambert J-C,

Carrasquillo MM, et al. Common variants at ABCA7, MS4A6A/

MS4A4E, EPHA1, CD33 and CD2AP are associated with Alz-

heimer’s disease. Nat Genet 2011;43:429–35.

[221] Guerreiro R, Wojtas A, Bras J, Carrasquillo M, Rogaeva E,

Majounie E, et al. TREM2 variants in Alzheimer’s disease. N Engl

J Med 2013;368:117–27.

[222] Corder EH, Saunders AM, Strittmatter WJ, Schmechel DE,

Gaskell PC, Small GW, et al. Gene dose of apolipoprotein E type 4

allele and the risk of Alzheimer’s disease in late onset families. Sci-

ence 1993;261:921–3.

[223] DeMattos RB, Cirrito JR, Parsadanian M, May PC, O’Dell MA,

Taylor JW, et al. ApoE and clusterin cooperatively suppress Abeta

levels and deposition: evidence that ApoE regulates extracellular

Abeta metabolism in vivo. Neuron 2004;41:193–202.

[224] Griciuc A, Serrano-Pozo A, Parrado AR, Lesinski AN, Asselin CN,

Mullin K, et al. Alzheimer’s disease risk gene CD33 inhibits micro-

glial uptake of amyloid beta. Neuron 2013;78:631–43.

[225] Satoh K, Abe-Dohmae S, Yokoyama S, St George-Hyslop P,

Fraser PE. ATP-binding cassette transporter A7 (ABCA7) loss of

function alters Alzheimer amyloid processing. J Biol Chem 2015;

290:24152–65.

[226] Sakae N, Liu C-C, ShinoharaM, Frisch-Daiello J, Ma L, Yamazaki Y,

et al. ABCA7 deficiency accelerates amyloid-b generation and Alz-

heimer’s neuronal pathology. J Neurosci 2016;36:3848–59.

[227] Poliani PL, Wang Y, Fontana E, Robinette ML, Yamanishi Y,

Gilfillan S, et al. TREM2 sustains microglial expansion during

aging and response to demyelination. J Clin Invest 2015;

125:2161–70.

[228] Ulrich JD, Holtzman DM. TREM2 function in Alzheimer’s disease

and neurodegeneration. ACS Chem Neurosci 2016;7:420–7.

[229] Bemiller SM, McCray TJ, Allan K, Formica SV, Xu G, Wilson G,

et al. TREM2 deficiency exacerbates tau pathology through dysregu-

lated kinase signaling in a mouse model of tauopathy. Mol Neurode-

gener 2017;12:74.

[230] Jay TR, Hirsch AM, Broihier ML, Miller CM, Neilson LE,

Ransohoff RM, et al. Disease progression-dependent effects of

TREM2 deficiency in a mouse model of Alzheimer’s disease. J Neu-

rosci 2017;37:637–47.

[231] Lyon KF, Strong CL, Schooler SG, Young RJ, Roy N, Ozar B, et al.

Natural variability of minimotifs in 1092 people indicates that mini-

motifs are targets of evolution. Nucleic Acids Res 2015;

43:6399–412.

[232] Roubroeks JAY, Smith RG, van den Hove DLA, Lunnon K. Epige-

netics and DNA methylomic profiling in Alzheimer’s disease and

other neurodegenerative diseases. J Neurochem 2017;143:158–70.

[233] Hwang J-Y, Aromolaran KA, Zukin RS. The emerging field of epige-

netics in neurodegeneration and neuroprotection. Nat Rev Neurosci

2017;18:347–61.

[234] Allis CD, Jenuwein T. Themolecular hallmarks of epigenetic control.

Nat Rev Genet 2016;17:487–500.

[235] Jimenez-Pacheco A, Franco JM, Lopez S, Gomez-Zumaquero JM,

Magdalena Leal-Lasarte M, Caballero-Hernandez DE, et al. Epige-

netic mechanisms of gene regulation in amyotrophic lateral sclerosis.

Adv Exp Med Biol 2017;978:255–75.

[236] Francelle L, Lotz C, Outeiro T, Brouillet E,Merienne K. Contribution

of neuroepigenetics to Huntington’s disease. Front Hum Neurosci

2017;11:17.

� 25 July 2018 � 9:16 am � ce

2253

Page 18: Minimotifs dysfunction is pervasive in neurodegenerative

23

S. Sharma et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions - (2018) 1-1918

2254

2255

2256

2257

2258

2259

2260

2261

2262

2263

2264

2265

2266

2267

2268

2269

2270

2271

2272

2273

2274

2275

2276

2277

2278

2279

2280

2281

2282

2283

2284

2285

2286

2287

2288

2289

2290

2291

2292

2293

2294

2295

2296

2297

2298

2299

2300

2301

2302

2303

2304

2305

2306

2307

2308

2309

2310

2311

2312

2313

2314

2315

2316

2317

2318

2319

2320

2321

2322

2323

2324

2325

2326

2327

2328

2329

2330

2331

2332

2333

2334

2335

2336

2337

2338

2339

2340

2341

2342

2343

2344

2345

2346

2347

2348

2349

2350

2351

2352

2353

2354

2355

2356

2357

2358

2359

2360

2361

2362

2363

2364

2365

2366

2367

2368

2369

2370

2371

2372

2373

2374

2375

2376

2377

2378

2379

2380

2381

2382

2383

2384

2385

2386

[237] Bassi S, Tripathi T, Monziani A, Di Leva F, Biagioli M. Epigenetics

of Huntington’s disease. Adv Exp Med Biol 2017;978:277–99.

[238] Narayan P, DragunowM. Alzheimer’s disease and histone code alter-

ations. Adv Exp Med Biol 2017;978:321–36.

[239] Pavlou MAS, Outeiro TF. Epigenetics in Parkinson’s disease. Adv

Exp Med Biol 2017;978:363–90.

[240] Liu H, Tang T-S, Guo C. Epigenetic profiles in polyglutamine disor-

ders. Epigenomics 2018;10:9–25.

[241] Usmani SS, Bedi G, Samuel JS, Singh S, Kalra S, Kumar P, et al.

THPdb: Database of FDA-approved peptide and protein therapeutics.

PLoS One 2017;12:e0181748.

[242] Caccamo A, Majumder S, Richardson A, Strong R, Oddo S. Molec-

ular interplay between mammalian target of rapamycin (mTOR),

amyloid-beta, and Tau: effects on cognitive impairments. J Biol

Chem 2010;285:13107–20.

[243] Spilman P, Podlutskaya N, Hart MJ, Debnath J, Gorostiza O,

Bredesen D, et al. Inhibition of mTOR by rapamycin abolishes cogni-

tive deficits and reduces amyloid-beta levels in a mouse model of

Alzheimer’s disease. PLoS One 2010;5:e9979.

[244] Rodriguez-Navarro JA, Cuervo AM. Autophagy and lipids: tight-

ening the knot. Semin Immunopathol 2010;32:343–53.

[245] Heiseke A, Aguib Y, Riemer C, Baier M, Sch€atzl HM. Lithium in-

duces clearance of protease resistant prion protein in prion-infected

cells by induction of autophagy. J Neurochem 2009;109:25–34.

[246] Webb JL, Ravikumar B, Atkins J, Skepper JN, Rubinsztein DC.

Alpha-Synuclein is degraded by both autophagy and the proteasome.

J Biol Chem 2003;278:25009–13.

[247] Ravikumar B, Duden R, Rubinsztein DC. Aggregate-prone proteins

with polyglutamine and polyalanine expansions are degraded by

autophagy. Hum Mol Genet 2002;11:1107–17.

[248] Skovronsky DM, Lee VM-Y, Trojanowski JQ. Neurodegenerative

diseases: new concepts of pathogenesis and their therapeutic implica-

tions. Annu Rev Pathol 2006;1:151–70.

[249] Rachakonda V, Pan TH, Le WD. Biomarkers of neurodegenerative

disorders: how good are they? Cell Res 2004;14:347–58.

[250] MacLeod R, Hillert E-K, Cameron RT, Baillie GS. The role and ther-

apeutic targeting of a-, b- and g-secretase in Alzheimer’s disease.

Future Sci OA 2015;1:FSO11.

[251] Evers MM, Toonen LJA, van Roon-MomWMC. Antisense oligonu-

cleotides in therapy for neurodegenerative disorders. Adv Drug Deliv

Rev 2015;87:90–103.

[252] Magen I, Hornstein E. Oligonucleotide-based therapy for neurode-

generative diseases. Brain Res 2014;1584:116–28.

[253] Gong B, Radulovic M, Figueiredo-Pereira ME, Cardozo C. The

ubiquitin-proteasome system: potential therapeutic targets for Alz-

heimer’s disease and spinal cord injury. Front Mol Neurosci 2016;

9:4.

[254] El-Agnaf OMA, Paleologou KE, Greer B, Abogrein AM, King JE,

Salem SA, et al. A strategy for designing inhibitors of alpha-

synuclein aggregation and toxicity as a novel treatment for Parkin-

son’s disease and related disorders. FASEB J 2004;18:1315–7.

[255] Schlessinger J, Lemmon MA. SH2 and PTB domains in tyrosine ki-

nase signaling. Sci STKE 2003;2003:RE12.

[256] Dev KK. Making protein interactions druggable: targeting PDZ do-

mains. Nat Rev Drug Discov 2004;3:1047–56.

[257] Abou-Abbass H, Abou-El-HassanH, BahmadH, Zibara K, Zebian A,

Youssef R, et al. Glycosylation and other PTMs alterations in neuro-

degenerative diseases: Current status and future role in neurotrauma.

Electrophoresis 2016;37:1549–61.

[258] Hawkes CA, Ng V, McLaurin J. Small molecule inhibitors of Ab-ag-

gregation and neurotoxicity. Drug Dev Res 2009;70:111–24.

[259] Heerboth S, Lapinska K, Snyder N, Leary M, Rollinson S, Sarkar S.

Use of epigenetic drugs in disease: an overview. Genet Epigenet

2014;6:9–19.

[260] Lewis S. Neurodegenerative disease: Restoring balance. Nat Rev

Neurosci 2016;17:400.

FLA 5.5.0 DTD � TRCI168_proof

[261] Bray N. Neurodegenerative disease: Losing the way. Nat Rev Neuro-

sci 2017;18:129.

[262] Krumova P, Weishaupt JH. Sumoylation in neurodegenerative dis-

eases. Cell Mol Life Sci 2013;70:2123–38.

[263] Oddo S. The ubiquitin-proteasome system in Alzheimer’s disease. J

Cell Mol Med 2008;12:363–73.

[264] D’Alton S, Lewis J. Therapeutic and diagnostic challenges for fron-

totemporal dementia. Front Aging Neurosci 2014;6:204.

[265] Sporns O, Chialvo DR, Kaiser M, Hilgetag CC. Organization, devel-

opment and function of complex brain networks. Trends Cogn Sci

2004;8:418–25.

[266] Zhang Q, Ma C, Gearing M, Wang PG, Chin L-S, Li L. Integrated

proteomics and network analysis identifies protein hubs and network

alterations in Alzheimer’s disease. Acta Neuropathol Commun 2018;

6:19.

[267] Crucitti P, Latora V, Marchiori M. Model for cascading failures in

complex networks. Phys Rev E Stat Nonlin Soft Matter Phys 2004;

69:045104.

[268] Turau V, Weyer C. Cascading failures caused by node overloading in

complex networks. IEEE 2016:1–6. Q

[269] Jones DT, Knopman DS, Gunter JL, Graff-Radford J, Vemuri P,

Boeve BF, et al. Cascading network failure across the Alzheimer’s

disease spectrum. Brain 2016;139:547–62.

[270] Jones DT, Graff-Radford J, Lowe VJ, Wiste HJ, Gunter JL,

SenjemML, et al. Tau, amyloid, and cascading network failure across

the Alzheimer’s disease spectrum. Cortex 2017;97:143–59.

[271] Gao H-M, Hong J-S. Why neurodegenerative diseases are progres-

sive: uncontrolled inflammation drives disease progression. Trends

Immunol 2008;29:357–65.

[272] Li Y, TanM-S, Jiang T, Tan L.Microglia in Alzheimer’s disease. Bio-

med Res Int 2014;2014:437483.

[273] Van Roey K, Gibson TJ, Davey NE.Motif switches: decision-making

in cell regulation. Curr Opin Struct Biol 2012;22:378–85.

[274] Alonso Vilatela ME, L�opez-L�opez M, Yescas-G�omez P. Genetics of

Alzheimer’s disease. Arch Med Res 2012;43:622–31.

[275] Thomas SN, Funk KE, Wan Y, Liao Z, Davies P, Kuret J, et al. Dual

modification of Alzheimer’s disease PHF-tau protein by lysine

methylation and ubiquitylation: a mass spectrometry approach.

Acta Neuropathol 2012;123:105–17.

[276] Bhattacharyya R, Barren C, Kovacs DM. Palmitoylation of amyloid

precursor protein regulates amyloidogenic processing in lipid rafts. J

Neurosci 2013;33:11169–83.

[277] Sacksteder CA, Qian W-J, Knyushko TV, Wang H, Chin MH,

Lacan G, et al. Endogenously nitrated proteins in mouse brain: links

to neurodegenerative disease. Biochemistry 2006;45:8009–22.

[278] Blokhuis AM, Groen EJN, Koppers M, van den Berg LH,

Pasterkamp RJ. Protein aggregation in amyotrophic lateral sclerosis.

Acta Neuropathol 2013;125:777–94.

[279] Lee S, Kim H-J. Prion-like mechanism in amyotrophic lateral scle-

rosis: are protein aggregates the key? Exp Neurobiol 2015;24:1–7.

[280] Furukawa Y, Kaneko K, Yamanaka K, O’Halloran TV, Nukina N.

Complete loss of post-translational modifications triggers fibrillar ag-

gregation of SOD1 in the familial form of amyotrophic lateral scle-

rosis. J Biol Chem 2008;283:24167–76.

[281] Valle C, Carr�ı MT. Cysteine modifications in the pathogenesis of

ALS. Front Mol Neurosci 2017;10:5.

[282] Freixes M, Puig B, Rodr�ıguez A, Torrej�on-Escribano B, Blanco R,

Ferrer I. Clusterin solubility and aggregation in Creutzfeldt-Jakob

disease. Acta Neuropathol 2004;108:295–301.

[283] Ermonval M, Mouillet-Richard S, Codogno P, Kellermann O, Botti J.

Evolving views in prion glycosylation: functional and pathological

implications. Biochimie 2003;85:33–45.

[284] Kaneko K, Vey M, Scott M, Pilkuhn S, Cohen FE, Prusiner SB.

COOH-terminal sequence of the cellular prion protein directs subcel-

lular trafficking and controls conversion into the scrapie isoform.

Proc Natl Acad Sci U S A 1997;94:2333–8.

� 25 July 2018 � 9:16 am � ce

2387

Page 19: Minimotifs dysfunction is pervasive in neurodegenerative

26

S. Sharma et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions - (2018) 1-19 19

2388

2389

2390

2391

2392

2393

2394

2395

2396

2397

2398

2399

2400

2401

2402

2403

2404

2405

2406

2407

2408

2409

2410

2411

2412

2413

2414

2415

2416

2417

2418

2419

2420

2421

2422

2423

2424

2425

2426

2427

2428

2429

2430

2431

2432

2433

2434

2435

2436

2437

2438

2439

2440

2441

2442

2443

2444

2445

2446

2447

2448

2449

2450

2451

2452

2453

2454

2455

2456

2457

2458

2459

2460

2461

2462

2463

2464

2465

2466

2467

2468

2469

2470

2471

2472

2473

2474

2475

2476

2477

2478

2479

2480

2481

2482

2483

[285] Gass CS, Luis CA, Meyers TL, Kuljis RO. Familial Creutzfeldt-

Jakob disease: a neuropsychological case study. Arch Clin Neuropsy-

chol 2000;15:165–75.

[286] Gambetti P, Kong Q, Zou W, Parchi P, Chen SG. Sporadic and famil-

ial CJD: classification and characterisation. Br Med Bull 2003;

66:213–39.

[287] Arrasate M, Finkbeiner S. Protein aggregates in Huntington’s dis-

ease. Exp Neurol 2012;238:1–11.

[288] Kumar KR, Weissbach A, HeldmannM, Kasten M, Tunc S, Sue CM,

et al. Frequency of the D620N mutation in VPS35 in Parkinson dis-

ease. Arch Neurol 2012;69:1360–4.

[289] Zimprich A, Benet-Pag�es A, Struhal W, Graf E, Eck SH, Offman MN,

et al. Amutation inVPS35, encoding a subunit of the retromer complex,

causes late-onset Parkinson disease.Am JHumGenet 2011;89:168–75.

[290] Vilari~no-G€uell C, Wider C, Ross OA, Dachsel JC, Kachergus JM,

Lincoln SJ, et al. VPS35 mutations in Parkinson disease. Am J

Hum Genet 2011;89:162–7.

[291] Wang W, Ma X, Zhou L, Liu J, Zhu X. A conserved retromer sorting

motif is essential for mitochondrial DLP1 recycling by VPS35 in Par-

kinson’s disease model. Hum Mol Genet 2017;26:781–9.

[292] Chartier-Harlin M-C, Dachsel JC, Vilari~no-G€uell C, Lincoln SJ,

Lepretre F, HulihanMM, et al. Translation initiator EIF4G1mutations

in familial Parkinson disease. Am J Hum Genet 2011;89:398–406.

[293] Cuervo AM, Stefanis L, Fredenburg R, Lansbury PT, Sulzer D.

Impaired degradation of mutant alpha-synuclein by chaperone-

mediated autophagy. Science 2004;305:1292–5.

[294] Vilari~no-G€uell C, Rajput A, Milnerwood AJ, Shah B, Szu-Tu C,

Trinh J, et al. DNAJC13 mutations in Parkinson disease. Hum Mol

Genet 2014;23:1794–801.

FLA 5.5.0 DTD � TRCI168_proof

[295] Bekris LM, Mata IF, Zabetian CP. The genetics of Parkinson disease.

J Geriatr Psychiatry Neurol 2010;23:228–42.

[296] Merry DE, Kobayashi Y, Bailey CK, Taye AA, Fischbeck KH. Cleav-

age, aggregation and toxicity of the expanded androgen receptor in

spinal and bulbar muscular atrophy. Hum Mol Genet 1998;

7:693–701.

[297] Panet-Raymond V, Gottlieb B, Beitel LK, Schipper H, Timiansky M,

Pinsky L, et al. Characterization of intracellular aggregates using

fluorescently-tagged polyglutamine-expanded androgen receptor.

Neurotox Res 2001;3:259–75.

[298] Berger TR Q, Montie HL, Jain P, Legleiter J, Merry DE. Identifi-

cation of novel polyglutamine-expanded aggregation species in

spinal and bulbar muscular atrophy. Brain Res 2015;1628:

254–64.

[299] Miller N, Feng Z, Edens BM, Yang B, Shi H, Sze CC, et al. Non-

aggregating tau phosphorylation by cyclin-dependent kinase 5 con-

tributes to motor neuron degeneration in spinal muscular atrophy. J

Neurosci 2015;35:6038–50.

[300] Mark MD, Krause M, Boele H-J, Kruse W, Pollok S, Kuner T,

et al. Spinocerebellar ataxia type 6 protein aggregates cause def-

icits in motor learning and cerebellar plasticity. J Neurosci 2015;

35:8882–95.

[301] Seki T, Shimahara T, Yamamoto K, Abe N, Amano T, Adachi N, et al.

Mutant gammaPKC found in spinocerebellar ataxia type 14 induces

aggregate-independent maldevelopment of dendrites in primary

cultured Purkinje cells. Neurobiol Dis 2009;33:260–73.

[302] Chen F, David D, Ferrari A, G€otz J. Posttranslational modifications of

tau–role in human tauopathies and modeling in transgenic animals.

Curr Drug Targets 2004;5:503–15.

� 25 July 2018 � 9:16 am � ce

2484

2485

2486

2487

2488

2489

2490

2491

2492

2493

2494

2495

2496

2497

2498

2499

2500

2501

2502

2503

2504

2505

2506

2507

2508

2509

2510

2511

2512

2513

2514

2515

2516

2517

2518

2519

2520

2521