Molecular genetics of early-onset Alzheimer's disease revisited

Molecular genetics of early-onset Alzheimer's disease revisited

Alzheimer’s & Dementia - (2016) 1-16 1 2 Review Article 3 4 5 6 7 a,b a,b a,b, 8Q10 a 9 Neurodegenerative Brain Diseases group, Department of Molecula...

2MB Sizes 0 Downloads 68 Views

Alzheimer’s & Dementia - (2016) 1-16 1 2 Review Article 3 4 5 6 7 a,b a,b a,b, 8Q10 a 9 Neurodegenerative Brain Diseases group, Department of Molecular Genetics, VIB, Antwerp, Belgium b 10Q1 Laboratory of Neurogenetics, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium 11 12 13 As the discovery of the Alzheimer disease (AD) genes, APP, PSEN1, and PSEN2, in families with 14Q2 Abstract autosomal dominant early-onset AD (EOAD), gene discovery in familial EOAD came more or less to 15 a standstill. Only 5% of EOAD patients are carrying a pathogenic mutation in one of the AD genes or 16 a APOE risk allele ε4, most of EOAD patients remain unexplained. Here, we aimed at summarizing 17 18 the current knowledge of EOAD genetics and its role in ongoing approaches to understand the 19 biology of AD and disease symptomatology as well as developing new therapeutics. Next, we 20 explored the possible molecular mechanisms that might underlie the missing genetic etiology of 21 EOAD and discussed how the use of massive parallel sequencing technologies triggered novel 22 gene discoveries. To conclude, we commented on the relevance of reinvestigating EOAD patients 23 as a means to explore potential new avenues for translational research and therapeutic discoveries. 24 Ó 2016 The Alzheimer’s Association. Published by Elsevier Inc. All rights reserved. 25 26 27 Keywords: Dementia; Early-onset Alzheimer disease; Genetics; Genome and exome sequencing; Missing genetic etiology; 28 Molecular pathways 29 30 31 32 33 with most of the EOAD patients being diagnosed between 1. Dementia and Alzheimer disease 34 45 and 60 years. Besides the typical clinical presentation 35 The term dementia is used to define a heterogeneous 36Q3 with memory impairment, atypical clinical presentation group of progressive and degenerative brain pathologies, 37 with focal cortical symptoms, for example, visual dysfuncclinically characterized by deterioration in memory, 38 tion, apraxia, dyscalculia, fluent and non-fluent aphasia, 39 learning, orientation, language, comprehension, and judgexecutive dysfunction, has also been reported. This atypical 40 ment. AD (OMIM# 104300), in its typical clinical presentapresentation is more frequently reported in EOAD patients 41 tion with progressive loss of memory and disturbance of 42 compared to LOAD, who mostly present with typical memadditional cognitive functions namely word-finding, spatial 43 ory phenotype [5]. Additionally, a nonmemory phenotype is cognition, reasoning, judgment, and problem solving [1], 44 seen in roughly 25% of EOAD patients in whom visual or 45 is the leading cause of dementia in the elderly. Of all demenapraxic and language phenotypes are more frequent [5]. 46 tia patients, 50% to 75% present with AD, which affects The neuropathologic hallmarks of AD brains are extra47 between 23 and 35 million people worldwide [2]. Age is 48 cellular accumulation of diffuse and neuritic amyloid the most prominent biological risk factor [3], and the age 49 plaques, composed of amyloid-b (Ab) peptide, and of 65 years is often used to classify AD patients in early50 frequently surrounded by dystrophic neurites and the intra51 onset (EOAD) and late-onset (LOAD) groups. Of all AD paneuronal accumulation of neurofibrillary tangles (NFTs) 52 tients, around 10% are diagnosed with EOAD [4], and they composed of hyperphosphorylated protein tau (p-tau) 53 present with their first symptoms between 30 and 65 years 54 [6,7]. These pathologic features are accompanied by

Molecular genetics of early-onset Alzheimer disease revisited Rita Cacace , Kristel Sleegers , Christine Van Broeckhoven

*Corresponding author. Tel.: 132 3 265 1101; Fax 132 3 265 1113. E-mail address: [email protected]

*

gliosis and the loss of neurons and synapses [7]. Although, some studies reported a larger neuropathologic burden [8] or a more widespread pathology extending outside the medial temporal lobe in younger patients [5], overall the

http://dx.doi.org/10.1016/j.jalz.2016.01.012 1552-5260/Ó 2016 The Alzheimer’s Association. Published by Elsevier Inc. All rights reserved. REV 5.4.0 DTD  JALZ2140_proof  23 March 2016  6:56 pm  ce

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

R. Cacace et al. / Alzheimer’s & Dementia - (2016) 1-16

2 110 111 112 113 114 115 116 117 118 119 120 121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176

pathologic features of EOAD and LOAD patients are largely similar, indicating that at the end-stage of disease, it is difficult to distinguish the two AD age groups by any other criterion than onset age. The main incentive for this review was the recent renewed interest in EOAD genetic studies due to the availability of high-throughput, genome-sequencing and exome-sequencing technologies, and bioinformatic tools, permitting new attempts to unravel the missing genetic etiology of EOAD. The expectations are that these new genetic approaches will uncover new molecular pathways or new molecular components of already known pathways. Furthermore, the availability of new genetic markers will help refining the different genetic signatures of clinical AD, allowing a more accurate stratification of patient cohorts, preclinical and clinical, for medical research, and for clinical trials. In the long term, the ability to identify different underlying molecular pathologies of clinical AD patients or at risk individuals will pave the way for personalized medicine and health care. 2. The genetic etiology of EOAD In contrast to LOAD which is a complex disorder with a heterogeneous etiology and an heritability of 70 to 80% [9,10], EOAD is an almost entirely genetically determined disease with a heritability ranging between 92% to 100% [9]. Between 35 to 60% of EOAD patients have at least one affected first-degree relative [11–13], and in 10% to 15% of those familial EOAD patients, the mode of inheritance is autosomal dominant transmission [11,13]. Genetic analysis of exceptionally large and informative monogenic pedigrees was the basis for the identification of high-penetrant mutations in the three EOAD genes, coding for the amyloid precursor protein (APP) and the presenilins 1 and 2 (PSEN1 and PSEN2). 2.1. Identification of causal EOAD genes in extended pedigrees Down syndrome (DS), caused by chromosome 21 (partial) trisomy, played a pivotal role in the early attempts to identify genes for inherited EOAD. DS patients were shown to present with a comparable brain pathology of amyloid plaques and tau tangles as AD patients [14]. The strong homology between the amyloid b (Ab) protein peptides, isolated from vessels [15] and from plaques [16] from DS and AD brains, was a first indication that both diseases shared a common genetic mechanism associated with chromosome 21 [15,17]. Whole-genome-linkage (WGL) studies in AD families provided supporting evidence for a genetic defect located on chromosome 21q [18–20]. Cloning of the gene coding for the amyloid b precursor protein (APP) [21], from which the amyloid b peptides are produced, and its mapping to chromosome 21q21.2–21q21.3 [22,23], encouraged a series of genetic studies aiming at

identifying mutations in AD patients and families. Initial genetic studies in large families with autosomal dominant AD were negative [24,25] but could be explained by the observation of a high degree of genetic heterogeneity in familial AD indicating that genes other than APP had to be involved [26]. A segregation study in extended multigenerational families with autosomal dominant cerebral hemorrhage with amyloidosis Dutch type (HCHWA-D), conclusively linked APP to the disease [27]. HCHWA-D brain pathology consists mainly of vascular amyloid depositions of the same Ab observed in AD brains [28]. Sequencing identified a mutation affecting the Ab sequence in patients with HCHWA-D [29] and segregated with disease [30]. The discovery of a APP mutation linked to vascular Ab pathology in HCHWA-D encouraged new mutation studies of APP in AD families. A first mutation was identified confirming a direct role for APP in AD pathogenesis in some AD families [31]. Segregation studies in EOAD pedigrees, negative for APP mutations, led to the identification of a new locus for EOAD on chromosome 14q24.3 [32–35]. Genetic mapping and gene cloning followed by mutation screening of candidate genes [36–38] identified presenilin 1 (PSEN1) as an EOAD gene with at that time unknown functions [39]. Based on protein homology, a second presenilin protein was identified and mapped to chromosome 1q31–q42 [40,41], in the region that was linked to AD in a series of families known as descendants of Volga-Germans [42,43] and was named presenilin 2 (PSEN2). 2.2. Genetic and phenotypic heterogeneity in EOAD To date, 52 pathogenic mutations in APP have been reported in 119 probands of autosomal dominant families (http://www.molgen.vib-ua.be/ADMutations) [44]. Most of the APP mutations are nonsynonymous within or flanking the Ab sequence (Fig. 1A). However, 25 genomic duplications of variable size containing APP have been identified co-segregating with AD in as many autosomal dominant families, as reported in http://www.molgen. vib-ua.be/ADMutations [44] and reviewed in [45], mimicking partial trisomy 21. Furthermore, a recessive one amino acid deletion (p.E693D) [46] and a recessive missense mutation with dominant negative effect on amyloidogenesis (p.A673V) [47,48] were described (Table 1). Q4 Missense mutations are identified at least 4-folds more frequently than APP genomic duplications in AD patients. Disease onset of APP mutation carriers ranged between 45 and 60 years [49,50]. In contrast to the missense mutations, showing a near-complete disease penetrance, APP genomic duplications display reduced penetrance and higher variability in onset age [45]. Besides the pathogenic mutations, a rare protective variant p.A673T in APP was reported that was enriched in the Icelandic population [51]. At the same amino acid position, the p.A673V variant showed

REV 5.4.0 DTD  JALZ2140_proof  23 March 2016  6:56 pm  ce

177 178 179 180 181 182 183 184 185 186 187 188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240 241 242 243

R. Cacace et al. / Alzheimer’s & Dementia - (2016) 1-16

web 4C=FPO

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

3

Fig. 1. APP protein mutations and structure. (A) APP protein sequence from amino acid residue 647 to 730 is presented, sequence in green depicts the extracellular domain, in orange the transmembrane domain and in dark blue the intracellular domain. Known pathogenic mutations are reported in purple and, if available, the mutation alias is shown, genomic duplications are not represented in the figure. In red are the two recessive pathogenic mutations, in gray the nonpathogenic mutations. A circle encloses the residue p.A673 because the change to T is described as protective against AD as well the change to V in the heterozygous state. An equal (5) marks a nonpathogenic silent mutation at residue p.G708. A delta (D) indicates a deletion. The cleavage sites of a, b, and g secretases are marked with black dotted lines. (B) Schematic presentation of the APP proteolytic processes. The nonamyloidogenic and the amyloidogenic pathways are illustrated. For clarity, a general g-secretase cleavage site is reported.

carriers have generally a wider onset age range from 40 to 70 years [49,50]. With a few exceptions, disease penetrance is complete for PSEN1 mutations [50]. In case of PSEN2 mutations, disease penetrance is more difficult to establish because far less families have been reported and the onset age range is much wider [50]. PSEN mutations are commonly inherited in an autosomal dominant manner, but de novo mutations in PSEN1 have been described in EOAD patients with disease onset as early as 28 years [52,53]. Two families have been described that segregate dominant EOAD mutations in a homozygous state, a Italian family with the APP mutation p.A713T [54] and a Colombian family with the PSEN1 mutation p.E280A [55]. In both families, the homozygous carriers did not seem to be more

antiamyloidogenic properties when identified in the heterozygous state [47], suggestive of a protective effect (Fig. 1A). Of all three EOAD genes, PSEN1 is most frequently mutated with 215 mutations (Fig. 2A) in 475 probands (http://www.molgen.vib-ua.be/ADMutations) [44]. In PSEN2, 31 mutations have been identified, 15 pathogenic in 24 probands and 16 with pathogenicity nature unclear (Fig. 2B). The mutation spectrum of the PSEN genes includes missense mutations, small insertions, and deletions (indels) as well as genomic deletions specifically in PSEN1, which lead to inframe exon 9 skipping (http://www.molgen.vib-ua.be/ ADMutations) [44] (Table 1). The onset age of carriers of a PSEN1 mutation ranged from 30 to 50 years. PSEN2 mutation

Table 1 Genetic heterogeneity in Alzheimer disease (AD): Known causal early-onset AD gene Gene

Chromosome

Inheritance

Gene identification

Mutation spectrum

Mutations (N)

APP

21q21.1–21q21.3

Autosomal dominant Autosomal recessive Protective

Linkage analysis

54*

PSEN1

14q24.3

Autosomal dominant de novo

Linkage analysis

PSEN2

1q31–q42

Autosomal dominant

Linkage and homology mapping

Missense Gene Duplication Amino acid deletion Missense Small indels Genomic deletions Missense

*The total number of APP mutations includes two causal recessive mutations. REV 5.4.0 DTD  JALZ2140_proof  23 March 2016  6:56 pm  ce

215

31

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

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

R. Cacace et al. / Alzheimer’s & Dementia - (2016) 1-16

web 4C=FPO

4

Fig. 2. PSENs protein mutations and structure. (A) PSEN1 and (B) PSEN2 protein sequences. In blue are marked the cytoplasmic domains, in yellow the transmembrane domains, in red the luminal domains, and in green the one intermembrane/IX transmembrane domain. Pathogenic or predicted pathogenic mutations are in purple. In orange are mutations with unclear pathogenicity and in gray are reported nonpathogenic mutations. Different nucleotide variants which lead to the same amino acid change, are reported only once. A delta (D) indicates deletions. An asterisk (*) marks two aspartates (D), which are the catalytic amino acids located in transmembrane domain VI (PSEN1 is D257 and PSEN2 is D263) and in transmembrane domain VII (PSEN1 is D385 and PSEN2 is D366). Arabic numbers indicate the last amino acid residue of every topological domain based on Uniprot database (PSEN1 P49768 and PSEN2 P49810). (C) Schematic presentation of APP and PSEN complexes. For PSENs, alternative predicted protein conformations are shown. The region with variable topology is framed in dotted line and includes either an intermembrane domain with intracellular C-terminus or a ninth transmembrane domain with extracellular C-terminus. The black dots on the transmembrane domains indicate the catalytic aspartates location. APP is inserted between PSENs transmembrane domains VI and VII. The a, b, and g secretases cleavage sites are schematically indicated.

severely affected [54,55]. The major implication of these findings is that the homozygous state of autosomal dominant mutations in AD is not lethal as initially hypothesized. This may also suggest the possible presence of additional protective genetic factors, which may modify the disease presentation. In an EOAD patient cohort, the estimated mutation frequencies for the three genes were ,1% for APP, 6% for PSEN1, and 1% for PSEN2 [56]. Together, they explain only 5%–10% of EOAD patients [9,56], whereas depending on the study, 23% to 88.2% of autosomal dominant patients remain genetically unexplained [9,11,57–59] (Fig. 3). The wide divergences in unexplained EOAD patients and families might result from differences in study design or sampling biases. Besides the genetic heterogeneity, phenotypic heterogeneity is also reported in EOAD. This complicates the clinical diagnosis in younger patients. Atypical presentation with language impairment was reported for specific PSEN1 mutations [49,50]. Prominent

behavioral symptoms (also presenting symptoms) like delusion, hallucinations, and apathy, are described for a subset of PSEN1 and PSEN2 mutations [49,50]. In some cases, the clinical symptoms fulfilled the criteria for a diagnosis of frontotemporal dementia, for example, in the case of the PSEN1 mutation p.G183V. In this specific example, the autoptic pathologic and immunohistochemistry analysis of the brain showed pick-type tauopathy, normally caused by MAPT mutations, in absence of extracellular Ab deposits [60]. The neurologic symptom of myoclonus is present in most of monogenic EOAD patients and increases with the disease duration; seizures were also reported as presenting symptom for some PSEN1 mutations and are very common in APP duplication carriers (including Down syndrome patients) [49,50]. Extrapyramidal symptoms are not uncommon in PSEN1 mutations carriers but very rare for PSEN2 and APP and tend to appear after several years into the disease course [49,50]. Spastic paraparesis has been associated with specific PSEN1

REV 5.4.0 DTD  JALZ2140_proof  23 March 2016  6:56 pm  ce

445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511

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

5

web 4C=FPO

R. Cacace et al. / Alzheimer’s & Dementia - (2016) 1-16

Fig. 3. Missing genetic etiology of early-onset Alzheimer disease (EOAD). The pie charts indicate the distribution of EOAD and late-onset Alzheimer disease (LOAD) patients (dark blue), the fraction of sporadic and familial EOAD patients with the sub-fraction of autosomal dominant patients (light emerald). The orange pie chart depicts the fraction of unexplained autosomal dominant families. The possible mechanisms that may explain the missing genetic etiology of EOAD are divided in two groups arising from the red pie chart: (1) possible undetected genetic alterations due to different causes, listed arising from the red pie chart (right side) and (2) possible undetected epigenetic dysregulation (left side). For both scenarios, some examples of study designs (e.g., family based, extreme trait design and so forth or investigation of DNA(de) methylation and so forth) and technological approaches such as next-generation sequencing (NGS), chromatin immunoprecipitation assay combined with sequencing (ChIP-Seq) and RNA sequencing (RNA-Seq) are schematically suggested.

mutations; in these cases, a peculiar pathologic finding of Ab plaques without a dense core or neuritic dystrophy (“cotton wool plaques”) is reported in the frontal cortex of the patients presenting the phenotype [61]. Cerebellar ataxia is a rare event but is present for specific PSEN1 mutations, like in the case of the p.S170F mutation co-segregating with a Cathepsin D variant, suggesting a deleterious epistatic effect on the disease course [62]. Taken together, this indicates that it is reasonable to continue efforts to both unravel the genetic etiology of EOAD and to search for modifier of the phenotype in presence of known pathogenic mutations. 2.3. APOE in EOAD A genome-wide linkage study in families with LOAD and subsequent association studies in patient/control cohorts

identified the ε4-allele of the apolipoprotein E gene (APOE) as a major genetic risk factor for LOAD [63–65]. The presence of one or two copies of the APOE ε4-allele increased the risk to develop LOAD by a factor 3 up to 15-fold in a dose-dependent manner [66,67]. The APOE ε4-allele also increased risk for EOAD in carriers of at least one ε4-allele and was highest in those with a positive family history (Table 2) [12]. In carriers, the homozygosity for the APOE ε4-allele was sufficient to significantly increase risk for EOAD independent of other genetic factors. In contrast, in carriers, heterozygous for the APOE ε4-allele risk was only significantly increased in the presence of a positive family history of disease, indicating that the presence of one ε4-allele was insufficient to increase risk for AD before the age of 65 years. It also suggested that the APOE ε4-allele may modify the expression of other genetic factors contributing to disease. However,

Table 2 Risk variants and genetic modifiers in EOAD Gene

Variant

Effect on EOAD

References

PSEN1 APOE

Promoter SNVs ε4-allele ε2-allele p.A23T p.M129V Octapeptide repeat insertions Nonsynonymous rare variants

Increased risk Increase risk—highest in the presence of positive family history—and earlier AAO Delays AAO Delays AAO Increased risk—VV homozygosity and highest in presence of positive family history Earlier AAO Increased risk—signal driven by positive family history

[68,69] [12,70,71] [70–73] [74] [75] [76] [77]

CCL11 PRNP SORL1

Abbreviations: EOAD, early-onset Alzheimer disease; SNVs, single nucleotide variants; AAO, age-at-onset. REV 5.4.0 DTD  JALZ2140_proof  23 March 2016  6:56 pm  ce

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

R. Cacace et al. / Alzheimer’s & Dementia - (2016) 1-16

6 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712

familial aggregation of EOAD cannot fully be explained by APOE as both carriers and noncarriers of a ε4-allele have an increased risk in the presence of a positive family history. Contrary to the risk-increasing effect of the APOE ε4allele, it was demonstrated that the ε2-allele exerted a protective effect [72,73]. Different from the mutations in APP, PSEN1, and PSEN2, the APOE ε4-allele was considered neither necessary nor sufficient to cause AD. However, this concept might need to be reconsidered based on a more recent study [78], which showed that the effect of APOE on AD risk is comparable to the effect of a major genetic factor with semidominant inheritance [78]. Analysis of the APOE genotypes in families segregating dominant APP (p.V717I) [79], PSEN1 (p.E280A) [70], or PSEN2 (p.N141I) mutations, showed that onset age decreased in presence of the ε4-allele in mutation carriers, whereas those with the ε2-allele had later onset ages [70,71]. A larger study of samples of the PSEN1 (p.E280A) family [73] confirmed the APOE ε2-allele as an age-at-onset (AAO) modifier (w12 years of delay) compared to the ε4-allele (Table 3) [73]. Nevertheless, these APOE genotype effects were not observed in all families with a pathogenic mutation [90]. Genome-wide searches in the Volga German families (PSEN2 p.N141I) identified at least three additional genetic loci (1q23.3, 17p13.2, and 7q33) [91], which could harbor modifier genes. Furthermore, a whole genome-sequencing approach of a large Colombian kindred (PSEN1 p.E280A), detected a protective variant (p.A23T, rs1129844) in the gene CCL11 encoding eotaxin-1 that delayed onset age of AD by a decade [74]. Autosomal dominant families, segregating mutations in one of the 3 causal EOAD genes, show often variability in clinical presentation, even in the presence of the same mutation. A meta-analysis study showed that the phenotypic heterogeneity could be partially explained by the mutated gene, the type of mutation, and the family history. But, this is not always the case, indicating that in certain families, additional modifier factors (i.e., gene-gene and geneenvironment interactions) may be involved [90]. 3. The amyloid cascade APP and its processed peptides are the cornerstone of the “amyloid cascade hypothesis” [92]. This hypothesis states that the accumulation of Ab is the causative agent of AD Table 3 Genetic heterogeneity in AD: Neurodegenerative brain diseases genes presenting with AD-like phenotype

Gene

Chromosome

Typical phenotype

GRN MAPT C9orf72 PRNP

17q21.31 17q21.31 9p21.2 20p13

FTLD FTLD FTLD Prion disease

Mutation frequency in AD (%)

References

,2% ,1% ,1% ,1%

[80–82] [83,84] [85–87] [88,89]

by destroying the synapses, inducing the formation of NFTs, and ultimately inducing neuronal loss [92]. Two distinct and mutually exclusive pathways cleave APP (Figure 1B). In one pathway, the cleavage by a-secretase and g-secretase produces three fragments: a secreted C-terminal fragment (sAPPa), p3, and the APP intracellular domain (AICD; Figure 1B). In the second, and thus amyloidogenic pathway, APP is cleaved by the b-secretase (beta-site APP cleaving enzyme 1, BACE1), followed by g-secretase cleavage. The cleavage by b-secretase generates a large soluble extracellular secreted domain (sAPPb). The remaining membrane bound APP fragment, C99, is processed by multiple g-secretase cleavages. The first occurs at the cleavage site epsilon (ε-site, Figure 1B) to produce the AICD and then, after g-secretase cleavages trim the membrane bound producing Ab species that differ in protein length including Ab38, Ab40 (most common fragment), Ab42 (self-aggregative fragment), and Ab43 [93]. Genetic mutations in APP are located in or near the Ab sequence and in the proximity of the cleavage sites of the secretases (Fig. 1A), whereas PSEN1 and PSEN2 mutations are scattered all over the protein (Fig. 2A and 2B). The mutations are predicted to cause AD through aberrant APP processing determining either increased Ab levels or increased production of Ab42 (and Ab43) peptide over Ab40, triggering Ab aggregation [93]. From its postulation, the “amyloid cascade hypothesis” has been supported and questioned (as reviewed by Morris et al. [94]) acknowledging that Ab has an important role in AD pathogenesis although its role in the disease process is most likely much more multifaceted. This was corroborated by the observation of a subgroup of clinically diagnosed AD patients with neurodegeneration and cognitive deficits but near absence of Ab deposits, defined as “suspected non-AD pathology” (SNAP) [95]. In a clinical trial of anti-Ab passive immunotherapy, 16.3% of the clinically diagnosed AD patients had a negative-baseline amyloid positron emission tomography scan [96]. Studies on mild cognitive impairment cohorts are ongoing to phenotypically classify the SNAP patients [97,98], to better understand, if they represent a subgroup of AD with pathologic features independent from the amyloid cascade [99]. 4. The role of other neurodegenerative brain diseases genes in EOAD expression Possibly, some of the SNAP patients might be associated with mutations in genes involved in other neurodegenerative brain diseases (NBD). Mutations have been identified, at low frequencies (less than 1%–2%), in genes causing frontotemporal lobar degeneration (FTLD), that is, mutations in MAPT [83,84] and in granulin (GRN) [80–82], and the repeat expansion in C9orf72 [85–87] (Table 3). The genetic heterogeneity of the phenotypic presentation of AD supports that both diseases form part of an AD-FTLD disease continuum [100,101]. An AD-like phenotype was also described in the presence of a nonsense mutation in the prion protein

REV 5.4.0 DTD  JALZ2140_proof  23 March 2016  6:56 pm  ce

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

R. Cacace et al. / Alzheimer’s & Dementia - (2016) 1-16 780 781 782 783 784 785 786 787 Q5 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

(PRNP p.Q160*) [88,89], gene responsible for inherited neurodegenerative spongiform encephalopathies. Histopathologic analysis of nonsense mutation carrier showed neuritic plaque-like pathology and severe NTFs, the plaques were immunonegative for Ab but immunopositive to PrP leading to a pathological diagnosis of prion disease [88] (Table 3). Moreover, a common coding polymorphism, M to V at position 129 of PRNP has been associated with EOAD when identified in homozygous state (MM and VV; Table 2) [75]. The risk was higher for the VV and increased in patients with positive family history [75]. In addition, increasing number of octapeptide repeat insertions in PRNP have also been associated with younger onset age in AD patients (Table 2) [76]. The genetic heterogeneity in some dementia patients leads to a later clinical diagnosis and to flawed genetic testing. With the current availability of massive parallel sequencing (MPS), causal genes across NBD clinical diagnostic dementia subgroups can be screened simultaneously, increasing the chances to identify pathologic mutations independent of the clinical diagnosis. It can be expected that the breadth and depth of this etiological disease heterogeneity will be clarified more fully in the coming years, as discussed in section 5 and 6. 5. The role of EOAD genes in AD susceptibility There is substantial evidence that apart from pathogenic mutations, the EOAD genes also harbor genetic variations that contribute to increased susceptibility for AD. For example, single nucleotide variants (SNVs) in the promoter region of APP or PSEN1 have been associated with increased risk of LOAD [68,69,102]. Some APP promotor SNVs, increase neuron-specific APP transcriptional activity by near two-fold [103,104], and as such increase risk to develop LOAD. PSEN1 promotor SNVs [68,69,102] could alter protein expression in neurons [69,102] leading to increased risk of both EOAD (Table 2) [68,69] and LOAD [69]. Likewise, coding variants in the known causal genes might influence AD risk; however, to date, there is little information available because causal EOAD genes are rarely screened in LOAD patients [56,105]. Moreover, in EOAD patients, the routine genetic testing paradigm was based on mutation frequency (PSEN1 . APP . PSEN2) and location of known mutations (exons 16 and 17 in APP covering the Ab peptide cleavages sites) and was stopped once a pathogenic mutation was identified. Today, the availability of extended gene panels and MPS technology, allows near unlimited screening of full coding sequences of multiple disease genes in parallel across clinical phenotype ([105,106] and unpublished data). Systematic screening of EOAD and LOAD patient cohorts showed rare or novel mutations in causal AD genes in LOAD patients ([105] and unpublished data) mutations in APP outside the Ab coding exons and the occurrence in one individual of mutations in more than one NBD genes (unpublished data). This suggests that mutations in known

7

EOAD genes might act differently on disease expression, which ranges from high penetrance (5 causal allele) and early-onset age to low penetrance (5 risk allele) and lateonset age, depending on the effect of the mutant allele on protein function ([105] and unpublished data). The new challenge that we are facing when using these gene panels is the high potential of identifying variants of uncertain significance (VUSs). These VUSs lack supportive functional data and can only be classified based on their likelihood of pathogenicity using in silico information obtained using bioinformatics prediction tools (e.g., predicting the impact of the mutated amino acid residue on protein stability/function) [107], data which are insufficient in a clinical diagnostic setting. 6. The missing genetics of EOAD Rare high-penetrant mutations in APP, PSEN1, and PSEN2 could explain only a small fraction of EOAD families, leaving a large group of autosomal dominant pedigrees genetically unexplained [9,11,57–59] (Fig. 3). Additionally, it was estimated that only between 5%–10% of EOAD [9,56] can be explained by mutations in the three known EOAD genes (Fig. 3). The limited number of resolved pedigrees and large number of genetically unexplained EOAD patients, indicated that additional causal genes remain to be identified. Next generation sequencing technologies (NGS), like whole-genome-sequencing (WGS) and wholeexome-sequencing (WES), offered a next step to gain new insights into the molecular genetic etiology of AD. Particularly, these NGS technologies made it conceivable to reconsider those small families that had been excluded from WGL studies because of their limited genetic information content and thus lack statistical power. Additionally, it became possible to examine, in an unbiased manner, groups of unrelated AD patients selected for their extreme phenotypic characteristics, for example, very early-onset age, familial history of disease, or atypical clinical and/or neuropathological phenotypes. An early NGS study produced WES data of a clinically diagnosed EOAD patient and identified in NOTCH3, a known mutation that had been associated with cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy [108], a dementia disorder that shares clinical similarities with EOAD. The index patient belonged to a consanguineous Turkish family with a complex clinical history of neurologic and immunologic disorders [108]. The study demonstrated that WES is a valid tool to unravel the genetic etiology of complex diseases [108]. A WES study in EOAD probands of autosomal dominant families, who were negative for mutations in the three EOAD genes, identified seven mutations on screening of 29 probands (five missense and two nonsense mutations) in the sorting protein-related receptor gene (SORL1) [109]. Previous studies have shown reduced expression of the SORL1 transcript in AD patients using

REV 5.4.0 DTD  JALZ2140_proof  23 March 2016  6:56 pm  ce

847 848 849 850 851 852 853 854 855 856 857 858 859 860 861 862 863 864 865 866 867 868 869 870 871 872 873 874 875 876 877 878 879 880 881 882 883 884 885 886 887 888 889 890 891 892 893 894 895 896 897 898 899 900 901 902 903 904 905 906 907 908 909 910 911 912 913

8 914 915 916 917 918 919 920 921 922 923 924 925 926 927 928 929 930 931 932 933 934 935 936 937 938 939 940 941 942 943 944 945 946 947 948 949 950 Q6 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

R. Cacace et al. / Alzheimer’s & Dementia - (2016) 1-16

microarray screening [110] and the involvement of the protein in APP and Ab trafficking [111]. One of the EOAD missense mutations in SORL1 (p.G511R) that cosegregates with autosomal dominant AD in one family [109], is located within the VPS10P domain of SORL1. This specific mutation was shown to reduce the capability of the VPS10P domain to bind Ab, resulting in accumulation of the peptide and lower turnover [112]. More, the investigation of an EOAD patient-control cohort confirmed the role of rare nonsynonymous coding variants in SORL1 as risk factor for EOAD (Table 2), with the association signal driven by familial patients [77]. Also, both common and rare, noncoding and coding variants in SORL1 have been associated with increased risk of LOAD [113–115]. Three SORL1 coding mutations (p.E270K and p.T947M, p.A528T) identified in a family-based and cohort-based association study on LOAD have been shown to increase Ab40 and/or Ab42 secretion when expressed in vitro [115]. These studies reinforced the role of rare variants in SORL1 in both EOAD and LOAD risk. The screening of SORL1 in larger patient/ control groups will help defining the contribution of rare genetic variants in SORL1 to AD etiology. 6.1. Causal genes in unexplained EOAD families Scarcity of sufficiently informative EOAD families, the small number of family members available for genetic analyses and the clinical and genetic heterogeneity, has hampered WGL studies in identifying the actual underlying gene defects [116]. An example is provided by the familybased GWL study in the Swedish series, which includes both EOAD and LOAD patients. The original WGL study in multiple autosomal dominant families detected a suggestive linkage at chromosome 5q35 [117]. The inclusion of additional families to the WGL analysis did not confirm the 5q35 locus [118], but the investigation of a selected number of families with autopsy confirmed AD revealed a suggestive locus on chromosome 8q24 [119]. An independent locus on chromosome 8q was identified in a Swedish family in another WGL study [120]. In the aforementioned examples, the underlying genetic defect has not yet been pinpointed. We also identified in one extended Dutch family, a significant WGL peak at chromosome 7q36 but candidate gene screening in this locus did not identify a mutation that could explain the linkage [121]. Today, NGS technologies are used to examine unexplained EOAD families [122]. In large extended families, the NGS approach can be preceded by a WGL analysis to prioritize chromosomal regions that are more likely to contain the disease gene. NGS data of two, distantly related, patients can be sufficient to identify shared, protein affecting, genetic variants in genes located in the linked loci. An advantage of this combined approach of WGL and NGS is a significant decrease in complexity of the NGS data under analysis. Furthermore, the success rate of

this approach can be substantially increased by including additional family members both affected and/or unaffected. The main strength of NGS technologies, however, is their capability to identify putative disease genes by direct analysis of the data obtained of related family members belonging to small nuclear families. Because of the familial nature, these NGS studies are often limiting their first analysis to variants in coding sequences, particularly variants predicted to have a high impact on the protein functioning and thus identifying high-penetrant causal mutations. But silent, noncoding and genomic variations can also exert an effect on disease risk or causality. For example, the intron 4 variant (c.358–304 C . G) in SOD1 defines the activation of a cryptic splice site with the inclusion of a pseudo-exon leading to amyotrophic lateral sclerosis [123]. Also, genomic duplications of the APP locus have been linked to EOAD (as reviewed in section 2.2), and an inversion polymorphism comprising MAPT has been associated to increased risk for neurodegenerative brain diseases like progressive supranuclear palsy [124,125]. In case, a candidate gene is identified in this NGS approach, the next major step is to generate confirmatory evidence of its pathogenic role in the disease. This can be obtained by cosegregation studies of the putative disease causing mutation in the family, by the screening of the candidate gene for mutations in a patient cohort and its absence in a control cohort. Limiting factors to the segregation analysis are the size of the pedigree as well as the number of available samples. Also, incomplete or age-dependent penetrance of the genetic variants can complicate the interpretation of both the segregation data as well as the presence of the variant in control individuals. Alternatively, an extreme trait design can be used to sequence a small number of individuals at one or both extremes of the disease (endo) phenotype [126] as shown by Nho et al. [127]. This approach, though it identified frequent variants that may be associated with AD risk, provides an example of using NGS with quantifiable traits as endophenotype. 6.2. Other mechanisms In most of the EOAD families, the disease inheritance is consistent with autosomal dominant transmission. Occasionally, other transmission patterns were reported like recessive mutations in APP [46,47] and de novo mutations in PSEN1 [52,53] (described in section 2.2), but they have not been systematically explored. A potential study design to optimize their detection is the use of WGS in small nuclear families or trios with neurologically healthy parents and EOAD patients negative for the APOE ε4-allele. It is possible to approach the data analysis, based on the family history of disease, to identify either recessive (positive family history) or de novo (negative family history) mutations. In specific cases, that is, when the family history is unknown, both hypotheses can be

REV 5.4.0 DTD  JALZ2140_proof  23 March 2016  6:56 pm  ce

981 982 983 984 985 986 987 988 989 990 991 992 993 994 995 996 997 998 999 1000 1001 1002 1003 1004 1005 1006 1007 1008 1009 1010 1011 1012 1013 1014 1015 1016 1017 1018 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

R. Cacace et al. / Alzheimer’s & Dementia - (2016) 1-16 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

followed in parallel. The WGS data of the affected offspring can be used to define a genome-wide map of runs of homozygosity (ROH), and these ROH regions can be analyzed together with the WGS data of the parents to identify recessive mutations. Recessive inheritance of AD has already been examined before using either polymorphic simple-tandem-repeat markers or high-density single nucleotide polymorphism genotyping arrays, identifying homozygous regions harboring potential candidate genes [128–131]. Conversely, the WGS data can be analyzed for de novo mutations present in the offspring but absent in the parents. Limitations to these study designs are the unavailability of both parents and, the possibility that EOAD in the offspring resulted from a dominant mutation that was undetected in the parent due to incomplete penetrance or an AAO modifier. Alternative to recessive or de novo mutations, sporadic EOAD patients can also be explained by germline or somatic mosaicism [132,133] as shown for the PSEN1 p.P436Q mutation [132], with the degree of mosaicism affecting onset age and clinical presentation. Also, epigenetic dysregulation can affect gene expression in AD. Deoxyribonucleic acid (DNA) (de)methylation, chromatin remodeling, and noncoding RNAs (ncRNAs) expression could play a more impacting role than initially thought in AD onset and expression (Fig. 3). Alteration in methylation in the promoter region of APP, PSEN1, and APOE have been described in AD patients as well as histone acetylation which was altered in both AD patients and in AD transgenic mouse models (as reviewed by Lardenoije et al., [134]). Imbalanced expression of noncoding RNAs, miRNA particularly, has been identified in relation to AD (as reviewed by van den Hove et al. [135]). These mechanisms affect gene expression without altering the DNA sequence and are currently underinvestigated in EOAD patients. Two epigenetic studies in LOAD [136,137], adopted novel designs to control for confounding factors like the different methylation pattern in different cells/tissues, interindividual variability within the same tissue [138]. Both studies independently detected changes in methylation in four CpG sites close to ANK1, RPL13, CDH23, and RHBDF2 [136,137]. This confirmed the respective findings and supported the strength of the study designs. Additionally, the recent availability of the epigenome roadmap [139], a public data resource containing epigenetic information from different cell types and tissues, is paving the way to more comprehensive research. This effort resulted, among others, in the identification of conserved epigenomic signals at orthologous regions between mice and humans, which upregulate the immune response genes and downregulate the synaptic plasticity genes in the CKp25 mouse model of AD [140]. The major challenge of this type of studies, in the years to come, will be to determine whether the epigenetic change is a cause or a consequence of the disease and how the disease risk is influenced by the alteration of the chromatin structure

9

[141]. Moreover, it will be relevant to comprehend how the epigenome signature varies between populations and how the epigenetic findings will translate from mouse models (e.g., CK-p25) to humans. The availability of epigenome data will allow the integration of genetic results with epigenome regulation to better understand disease signatures which may represent a potential target for future diagnostic and treatment interventions. 7. Translational research in autosomal dominant families On the one hand, the investigation of unexplained EOAD families and patients aims at identifying novel causal genes and pathways. On the other hand, an international effort is ongoing to better understand the temporal sequence and the earliest events, which lead to the endpoint disease clinical manifestation. Longitudinal studies on families segregating a monogenic form of AD and patients with a known causal mutation are currently performed to investigate the prodromal phase of the neurodegenerative processes which ultimately lead to AD [142]. The understanding of the post-genomic consequences of the genetic mutations aims at improving the AD diagnosis through the identification of novel-sensitive biomarkers (Box 1), which correlate with earliest pathologic events and with disease conversion and progression. This will also help in the definition of targets for therapeutic screening and the selection of uniform patients groups for targeted pharmaceutical approaches. Longitudinal studies in asymptomatic mutation carriers have already demonstrated that at least 20 years before the clinical manifestation of cognitive deficits it is possible to detect the pathologic changes happening in the brain using both biochemical and instrumental biomarkers (Box 1) [142,144]. The longitudinal data investigation of cognitively unimpaired mutations carriers from the Colombian PSEN1 kindred included in the Alzheimer’s Prevention Initiative (API), for example, led to the identification of a composite cognitive test score that has improved power to detect preclinical AD decline [150]. A direct application of this score could be in identifying preclinical patients to include in prevention trials [150]. In the API cohort, the anti-Ab monoclonal antibody crenezumab testing is ongoing [151]. Another example of translational research is provided by the dominantly inherited Alzheimer’s network trials unit, which is currently testing in presymptomatic known mutation carriers two monoclonal anti-Ab antibodies (gantenerumab and solanezumab) that target different Ab species [151]. Ab42-reducing approaches may have more efficacy in familial patients, because the increased production of Ab42 is caused by genetic mutations, in this way, it is possible to attempt to increase the clearance of the toxic peptide before the accumulation is too extensive. Taken together, these examples demonstrate that families segregating a monogenic form of AD and patients with a known causal

REV 5.4.0 DTD  JALZ2140_proof  23 March 2016  6:56 pm  ce

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

10 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

R. Cacace et al. / Alzheimer’s & Dementia - (2016) 1-16

Box 1 Genetic, biochemical, neuroanatomic, and metabolic markers in AD diagnosis

Genetic markers: APP, PSEN1, and PSEN2 APP (OMIM# 104760) contains 18 exons and spans a genomic region of 6290 kb at chromosome 21q21.1–21q21.3. APP has several isoforms generated by alternative splicing. The APP770 is used as canonical isoform for mutation nomenclature. Reference sequences are Gene NG_007376.1, cDNA NM_000484.2, and Protein NP_000475.1. The Ab protein is encoded by exons 16 and 17. APP is a type I transmembrane protein (Figure 1). PSEN1 (OMIM# 104311) counts 13 exons and spans 6 84 kb at chromosome 14q24.3. The first 4 exons contain untranslated sequence, and exons 1 and 2 represent alternate transcription initiation sites. PSEN1 counts 467 amino acids residues. Reference sequences are Gene NG_007386.2, cDNA NM_000021. 2, and protein NP_000012.1. PSEN2 (OMIM# 600759) contains 12 exons and spans a region of 625 kb at chromosome 1q42.13. The first two exons encode the 5-prime untranslated region. PSEN2 has 448 amino acids. Reference sequences are Gene NG_007381.1, cDNA NM_000447.1, and protein NP_000438.1. The PSENs share an overall amino acid homology of 67% and are multimeric protein predicted to cross the membrane 7–9 times (Fig. 2C). Diagnostic molecular genetic screenings have been performed so far by sequencing exon 16 and 17 (Ab coding exons) of APP and all coding exons of PSEN1 and PSEN2, after a decision paradigm based on gene mutation frequency PSEN1 . APP . PSEN2. NGS technologies are sequencing in parallel the entire coding sequence of all three causal genes either by gene-panels approach [106] or by whole-exome sequencing [143]. Specific (or core) biochemical markers are Ab42, total tau (t-tau), and phosphorylated tau (p-tau). These are dosed in cerebrospinal fluid (CSF) on lumbar puncture. These biomarkers are measured to provide a reflection of the cerebral metabolic process of the protein transport between the brain and the CSF. General consensus is that in AD patients, the CSF levels of Ab42 are reduced, because of amyloid plaque formation in the brain, and that t-tau and p-tau are increased because of neuronal cell loss [144,145]. Furthermore, t-tau CSF levels increase with AD progression [145]. The better understanding of the pathologic processes occurring in AD brains are triggering the validation of novel CSF biomarkers (as reviewed in [146]) as the APP-cleaving enzyme 1 (BACE1) levels, the dosage of truncated amyloid-b isoforms, both linked to amyloidogenesis and result increased in patients. APP cleavage products, which are produced during APP processing can be measured to assess drug efficacy.

F2-isoprostane a marker of mithocandrial dysfunction as well as markers of synaptic degeneration (e.g., synaptotagmin, synapsin, synaptophysin and so forth) and markers of brain injury (i.e., visinin-like protein 1 (VILIP-1) [147]), are under investigation to assess their applicability in diagnostic. Neuroanatomic markers such as computer-assisted tomography (CT) and volumetric magnetic resonance imaging (MRI) are used to measure structural brain atrophy. Alteration in brain metabolic processed are measured using metabolic markers, that is, positronemission tomography and single photon-emission computed tomography (SPECT) with radiopharmaceutical agents, for example, fluorodeoxyglucose (FDG). It is also possible to image in vivo amyloid deposition, imaged by PET with the use of Pittsburgh compound B (PiB). A recent study shows that also tau tangles correlate well with AD diagnosis, showing the strongest predictive value for progression to AD [148]. Although tau is a biomarker of brain injury, synapse loss, and progression to neurodegeneration, selective tau tracers for PET are not yet available [149]. In vivo tau imaging will help in clarifying the role of tau in AD onset and progression and will support the clinical disease diagnosis [149]. mutation are still in the central core of preclinical and clinical research. The knowledge of the molecular and cellular biology of AD has already supported the development of tools for disease diagnosis (Box 1), but efforts are still ongoing to identify novel biomarkers for disease prediction and prognosis. These will allow the selection of homogeneous cohorts of participants for clinical trials with a close monitoring of the drug response.

8. Lesson learned from research in LOAD Locus heterogeneity and limitations in both pedigree size and technologies were probably the major contributors to the lack of additional genetic breakthroughs in EOAD. Meanwhile, the investigation of LOAD, using genome-wide association studies (GWAS), led to the identification of common risk variants within .20 genetic loci [114,152–156]. These genetic loci could be grouped in three major biological pathways: immune system; lipid metabolism; and synaptic dysfunction/cell membrane processes (e.g., endocytosis) [116,157,158]. Additionally, NGS resequencing efforts are identifying rare variants (minor allele frequency ,1% in the general population) with a strong effect on disease risk in additional genes or in genes previously associated with LOAD. Examples are provided by rare variants detected in TREM2 [159,160] or ABCA7 [161,162]. Also, in these cases, the main pathways involved are the immune response (TREM2, ABCA7) and lipid metabolism (ABCA7) [157]. As

REV 5.4.0 DTD  JALZ2140_proof  23 March 2016  6:56 pm  ce

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

R. Cacace et al. / Alzheimer’s & Dementia - (2016) 1-16 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

for APOE ε4 (discussed in section 2.3), a few rare variants in the risk genes (e.g., TREM2 [163], ABCA7 [162], EPHA1 [164]) have been shown segregating in families, suggesting a possible effect on disease penetrance, although further studies are needed to strengthen this hypothesis. The role of rare variants in EOAD has been less investigated nonetheless a few examples are available like the rare protective variant detected in APP [51] and the variant in CCL11 (eotaxin-1) [74]. This last example is of particular relevance, as it demonstrated an involvement of the immune pathway in modulating the disease onset in presence of a highly penetrant mutation in PSEN1. The molecular mechanism through which eotaxin-1 modulates AD onset needs further investigation but, if validated, may represent a first step to develop therapies able to delay the onset of AD. This finding also supports the hypothesis that rare variants can modulate the disease onset and most likely also the disease progression. Taken together, the findings suggest that Ab processing in LOAD could have a lesser central role, whereas the clearance of the aggregates might be involved in the disease pathogenesis [165]. On the one hand, the identification in EOAD of AAO disease modifiers (APOE: lipid metabolism and CCL11: immune pathway) in genes belonging to two of the biological pathways detected by GWAS in LOAD, is indicative of multiple (common) pathways modulating the clinical expression of AD. On the other hand, the shared clinical features as well as the pathologic findings in brain tissue of both EOAD and LOAD suggest that discoveries obtained in EOAD will likely translate to LOAD.

11

progresses have been made in the last years in disclosing the pathologic mechanisms of AD. Novel regulatory mechanism and biological pathways have been disclosed, but prevention of the disease is still a challenge ahead. In this review, we have summarized the main aspects of the research in EOAD and how these have influenced the current knowledge of the disease mechanisms. We suggest that continuation of the investigations of families segregating known mutations and the elucidation of the missing genetic etiology in unexplained EOAD patients still has a vast potential to deliver novel crucial pieces that will lead to a better understanding of the complex puzzle of AD at large. Acknowledgments The research in the authors’ group is funded in part by a MetLife Foundation Award for Medical Research (USA), a U.S. Army Medical Research and Material Command (USAMRMC) Research Award, a Janssen Pharmaceutica Stellar Research Project, the Belgian Science Policy Office Interuniversity Attraction Poles program, the Alzheimer Research Foundation (SAO-FRA), the Flemish Government initiated Flanders Impulse Program on Networks for Dementia Research (VIND), the Flemish Government initiated Methusalem Excellence Program, the Research Foundation Flanders (FWO), and the University of Antwerp Research Fund, Belgium.

RESEARCH IN CONTEXT 9. Conclusions The renewed interest in EOAD in the NGS era has the potential to implement the knowledge of the molecular and cellular mechanisms which ultimately lead to AD. The indepth genetic characterization, by systematic screening of the dementia causal genes, will allow patient stratification in more homogenous groups, leading to the selection of unexplained EOAD, both familiar and sporadic patients to include in further research. This can be gene hunting projects or trials for biomarkers selection or compound testing, paving the way to personalized medicine. In parallel to the deep genetic profiling of patients cohorts, the longitudinal follow-up of families with known mutations will provide additional insights in the known disease process that could have implications for diagnostics, for the development of predictive tests, and for clinical trial design. In a future prospective, the integration of NGS with data coming from other–omics analysis (i.e., epigenomics, proteomics, transcriptomics, and metabolomics), might lead to the identification of molecular disease signatures, identifying pattern(s) of altered protein expression that are better able to discriminate early disease stages and facilitate the identification of novel causal proteins and pathways or to prioritize genes and proteins as “druggable” molecular targets for novel therapeutic approaches. In conclusion, tremendous

1. Systematic review: PubMed search, meeting abstracts and presentations, were used to collect information concerning Alzheimer disease (AD). The search focus was on the genetics of early-onset AD (EOAD), on the application of high-throughput sequencing technologies and on ongoing translational studies on EOAD. 2. Interpretation: We provide a comprehensive review on EOAD genetics. Despite the tremendous advance in the field, a large part of the EOAD patients is still unexplained. Thus, possible molecular mechanisms that might underlie the missing genetic etiology of EOAD are explored. 3. Future directions: The basis of the current knowledge of AD is derived from genetic studies on large EOAD pedigrees in the early 90s. Here, we suggest that the use of high-throughput sequencing technologies applied to EOAD patients has a vast potential to deliver crucial information to help in better understanding the molecular mechanisms of AD in general.

REV 5.4.0 DTD  JALZ2140_proof  23 March 2016  6:56 pm  ce

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

12 1450 1451 1452 1453 1454 1455 1456 1457 1458 1459 Q7 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

R. Cacace et al. / Alzheimer’s & Dementia - (2016) 1-16

References [1] McKhann GM, Knopman DS, Chertkow H, Hyman BT, Jack CR Jr, Kawas CH, et al. The diagnosis of dementia due to Alzheimer’s disease: recommendations from the National Institute on AgingAlzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement 2011;7:263–9. [2] Alzheimer’s Disease International. World Alzheimer Report 2015: The global impact of dementia an analysis of prevalence, incidence, costs and trends; 2015. [3] Carr DB, Goate A, Phil D, Morris JC. Current concepts in the pathogenesis of Alzheimer’s disease. Am J Med 1997;103:3S–10. [4] Alzheimer’s Disease International. World Alzheimer Report 2009: The Global Prevalence of Dementia; 2009;. p. 1–96. [5] van der Flier WM, Pijnenburg YA, Fox NC, Scheltens P. Early-onset versus late-onset Alzheimer’s disease: the case of the missing APOE varepsilon4 allele. Lancet Neurol 2011;10:280–8. [6] Braak H, Braak E. Demonstration of amyloid deposits and neurofibrillary changes in whole brain sections. Brain Pathol 1991;1:213–6. [7] Hyman BT, Phelps CH, Beach TG, Bigio EH, Cairns NJ, Carrillo MC, et al. National Institute on Aging-Alzheimer’s Association guidelines for the neuropathologic assessment of Alzheimer’s disease. Alzheimers Dement 2012;8:1–13. [8] Marshall GA, Fairbanks LA, Tekin S, Vinters HV, Cummings JL. Early-onset Alzheimer’s disease is associated with greater pathologic burden. J Geriatr Psychiatry Neurol 2007;20:29–33. [9] Wingo TS, Lah JJ, Levey AI, Cutler DJ. Autosomal recessive causes likely in early-onset Alzheimer disease. Arch Neurol 2012; 69:59–64. [10] Gatz M, Reynolds CA, Fratiglioni L, Johansson B, Mortimer JA, Berg S, et al. Role of genes and environments for explaining Alzheimer disease. Arch Gen Psychiatry 2006;63:168–74. [11] Jarmolowicz AI, Chen HY, Panegyres PK. The Patterns of Inheritance in Early-Onset Dementia: Alzheimer’s Disease and Frontotemporal Dementia. Am J Alzheimers Dis Other Demen 2014; 30:299–306. [12] van Duijn CM, de Knijff P, Cruts M, Wehnert A, Havekes LM, Hofman A, et al. Apolipoprotein E4 allele in a population-based study of early-onset Alzheimer’s disease. Nat Genet 1994;7:74–8. [13] Campion D, Dumanchin C, Hannequin D, Dubois B, Belliard S, Puel M, et al. Early-onset autosomal dominant Alzheimer disease: prevalence, genetic heterogeneity, and mutation spectrum. Am J Hum Genet 1999;65:664–70. [14] Wisniewski KE, Dalton AJ, McLachlan C, Wen GY, Wisniewski HM. Alzheimer’s disease in Down’s syndrome: clinicopathologic studies. Neurology 1985;35:957–61. [15] Glenner GG, Wong CW. Alzheimer’s disease and Down’s syndrome: sharing of a unique cerebrovascular amyloid fibril protein. Biochem Biophys Res Commun 1984;122:1131–5. [16] Masters CL, Simms G, Weinman NA, Multhaup G, McDonald BL, Beyreuther K. Amyloid plaque core protein in Alzheimer disease and Down syndrome. Proc Natl Acad Sci U S A 1985;82:4245–9. [17] Glenner GG, Wong CW. Alzheimer’s disease: initial report of the purification and characterization of a novel cerebrovascular amyloid protein. Biochem Biophys Res Commun 1984;120:885–90. [18] George-Hyslop PH, Tanzi RE, Polinsky RJ, Haines JL, Nee L, Watkins PC, et al. The genetic defect causing familial Alzheimer’s disease maps on chromosome 21. Science 1987;235:885–90. [19] Goate AM, Haynes AR, Owen MJ, Farrall M, James LA, Lai LY, et al. Predisposing locus for Alzheimer’s disease on chromosome 21. Lancet 1989;1:352–5. [20] Hardy J, Goate A, Owen M, Rossor M. Presenile dementia associated with mosaic trisomy 21 in a patient with a Down syndrome child. Lancet 1989;2:743. [21] Kang J, Lemaire HG, Unterbeck A, Salbaum JM, Masters CL, Grzeschik KH, et al. The precursor of Alzheimer’s disease amyloid

[22]

[23]

[24]

[25]

[26]

[27]

[28]

[29]

[30]

[31]

[32]

[33]

[34]

[35]

[36]

[37]

[38]

A4 protein resembles a cell-surface receptor. Nature 1987; 325:733–6. Goldgaber D, Lerman MI, McBride OW, Saffiotti U, Gajdusek DC. Characterization and chromosomal localization of a cDNA encoding brain amyloid of Alzheimer’s disease. Science 1987;235:877–80. Tanzi RE, Gusella JF, Watkins PC, Bruns GA, George-Hyslop P, Van Keuren ML, et al. Amyloid beta protein gene: cDNA, mRNA distribution, and genetic linkage near the Alzheimer locus. Science 1987; 235:880–4. Van Broeckhoven C, Genthe AM, Vandenberghe A, Horsthemke B, Backhovens H, Raeymaekers P, et al. Failure of familial Alzheimer’s disease to segregate with the A4- amyloid gene in several European families. Nature 1987;329:153–5. Tanzi RE, George-Hyslop PH, Haines JL, Polinsky RJ, Nee L, Foncin JF, et al. The genetic defect in familial Alzheimer’s disease is not tightly linked to the amyloid beta-protein gene. Nature 1987; 329:156–7. St George-Hyslop PH, Haines JL, Farrer LA, Polinsky R, Van Broeckhoven C, Goate A, et al. Genetic linkage studies suggest that Alzheimer’s disease is not a single homogeneous disorder. Nature 1990;347:194–7. Van Broeckhoven C, Haan J, Bakker E, Hardy JA, Van Hul W, Wehnert A, et al. Amyloid beta protein precursor gene and hereditary cerebral hemorrhage with amyloidosis (Dutch). Science 1990; 248:1120–2. Coria F, Prelli F, Castano EM, Larrondo-Lillo M, FernandezGonzalez J, van Duinen SG, et al. Beta-protein deposition: a pathogenetic link between Alzheimer’s disease and cerebral amyloid angiopathies. Brain Res 1988;463:187–91. Levy E, Carman MD, Fernandez-Madrid IJ, Power MD, Lieberburg I, van Duinen SG, et al. Mutation of the Alzheimer’s disease amyloid gene in hereditary cerebral hemorrhage, Dutch type. Science 1990; 248:1124–6. Bakker E, Van Broeckhoven C, Haan J, Voorhoeve E, Van Hul W, Levy E, et al. DNA diagnosis for hereditary cerebral hemorrhage with amyloidosis (Dutch type). Am J Hum Genet 1991;49:518–21. Goate A, Chartier-Harlin MC, Mullan M, Brown J, Crawford F, Fidani L, et al. Segregation of a missense mutation in the amyloid precursor protein gene with familial Alzheimer’s disease. Nature 1991;349:704–6. Schellenberg GD, Bird TD, Wijsman EM, Orr HT, Anderson L, Nemens E, et al. Genetic linkage evidence for a familial Alzheimer’s disease locus on chromosome 14. Science 1992;258:668–71. George-Hyslop PH, Haines J, Rogaev E, Mortilla M, Vaula G, Pericak-Vance M, et al. Genetic evidence for a novel familial Alzheimer’s disease locus on chromosome 14. Nat Genet 1992;2:330–4. Van Broeckhoven C, Backhovens H, Cruts M, De Winter G, Bruyland M, Cras P, et al. Mapping of a gene predisposing to early-onset Alzheimer’s disease to chromosome 14q24.3. Nat Genet 1992;2:335–9. Mullan M, Houlden H, Windelspecht M, Fidani L, Lombardi C, Diaz P, et al. A locus for familial early-onset Alzheimer’s disease on the long arm of chromosome 14, proximal to the alpha 1antichymotrypsin gene. Nat Genet 1992;2:340–2. Clark RF, Cruts M, Korenblat KM, He C, Talbot C, Van Broeckhoven C, et al. A yeast artificial chromosome contig from human chromosome 14q24 spanning the Alzheimer’s disease locus AD3. Hum Mol Genet 1995;4:1347–54. Mitsunaga Y, Takahashi K, Tabira T, Tasaki H, Watanabe S. Assignment of a familial Alzheimer’s disease locus between D14S289 and D14S53. Lancet 1994;344:1154–5. Cruts M, Backhovens H, Theuns J, Clark RF, Le Paslier D, Weissenbach J, et al. Genetic and physical characterization of the early-onset Alzheimer’s disease AD3 locus on chromosome 14q24.3. Hum Mol Genet 1995;4:1355–64.

REV 5.4.0 DTD  JALZ2140_proof  23 March 2016  6:56 pm  ce

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 1551 1552 1553 1554 1555 1556 1557 1558 1559 1560 1561 1562 1563 1564 1565 1566 1567 1568 1569 1570 1571 1572 1573 1574 1575 1576 1577 1578 1579 1580 1581 1582 1583

R. Cacace et al. / Alzheimer’s & Dementia - (2016) 1-16 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

[39] Sherrington R, Rogaev EI, Liang Y, Rogaeva EA, Levesque G, Ikeda M, et al. Cloning of a gene bearing missense mutations in early-onset familial Alzheimer’s disease. Nature 1995;375:754–60. [40] Rogaev EI, Sherrington R, Rogaeva EA, Levesque G, Ikeda M, Liang Y, et al. Familial Alzheimer’s disease in kindreds with missense mutations in a gene on chromosome 1 related to the Alzheimer’s disease type 3 gene. Nature 1995;376:775–8. [41] Levy-Lahad E, Wasco W, Poorkaj P, Romano DM, Oshima J, Pettingell WH, et al. Candidate gene for the chromosome 1 familial Alzheimer’s disease locus. Science 1995;269:973–7. [42] Bird TD, Lampe TH, Nemens EJ, Miner GW, Sumi SM, Schellenberg GD. Familial Alzheimer’s disease in American descendants of the Volga Germans: probable genetic founder effect. Ann Neurol 1988;23:25–31. [43] Levy-Lahad E, Wijsman EM, Nemens E, Anderson L, Goddard KA, Weber JL, et al. A familial Alzheimer’s disease locus on chromosome 1. Science 1995;269:970–3. [44] Cruts M, Theuns J, Van Broeckhoven C. Locus-specific mutation databases for neurodegenerative brain diseases. Hum Mutat 2012; 33:1340–4. [45] Hooli BV, Mohapatra G, Mattheisen M, Parrado AR, Roehr JT, Shen Y, et al. Role of common and rare APP DNA sequence variants in Alzheimer disease. Neurology 2012;78:1250–7. [46] Tomiyama T, Nagata T, Shimada H, Teraoka R, Fukushima A, Kanemitsu H, et al. A new amyloid beta variant favoring oligomerization in Alzheimer’s-type dementia. Ann Neurol 2008;63:377–87. [47] Di Fede G, Catania M, Morbin M, Rossi G, Suardi S, Mazzoleni G, et al. A recessive mutation in the APP gene with dominant-negative effect on amyloidogenesis. Science 2009;323:1473–7. [48] Giaccone G, Morbin M, Moda F, Botta M, Mazzoleni G, Uggetti A, et al. Neuropathology of the recessive A673V APP mutation: Alzheimer disease with distinctive features. Acta Neuropathol 2010; 120:803–12. [49] Ryan NS, Rossor MN. Correlating familial Alzheimer’s disease gene mutations with clinical phenotype. Biomark Med 2010;4:99–112. [50] Pilotto A, Padovani A, Borroni B. Clinical, biological, and imaging features of monogenic Alzheimer’s Disease. Biomed Res Int 2013; 2013:689591. [51] Jonsson T, Atwal JK, Steinberg S, Snaedal J, Jonsson PV, Bjornsson S, et al. A mutation in APP protects against Alzheimer’s disease and age-related cognitive decline. Nature 2012;488:96–9. [52] Portet F, Dauvilliers Y, Campion D, Raux G, Hauw JJ, Lyon-Caen O, et al. Very early onset AD with a de novo mutation in the presenilin 1 gene (Met 233 Leu). Neurology 2003;61:1136–7. [53] Golan MP, Styczynska M, Jozwiak K, Walecki J, Maruszak A, Pniewski J, et al. Early-onset Alzheimer’s disease with a de novo mutation in the presenilin 1 gene. Exp Neurol 2007;208:264–8. [54] Conidi ME, Bernardi L, Puccio G, Smirne N, Muraca MG, Curcio SA, et al. Homozygous carriers of APP A713T mutation in an autosomal dominant Alzheimer disease family. Neurology 2015; 84:2266–73. [55] Kosik KS, Munoz C, Lopez L, Arcila ML, Garcia G, Madrigal L, et al. Homozygosity of the autosomal dominant Alzheimer disease presenilin 1 E280A mutation. Neurology 2015;84:206–8. [56] Brouwers N, Sleegers K, Van Broeckhoven C. Molecular genetics of Alzheimer’s disease: an update. Ann Med 2008;40:562–83. [57] Wallon D, Rousseau S, Rovelet-Lecrux A, Quillard-Muraine M, Guyant-Marechal L, Martinaud O, et al. The French series of autosomal dominant early onset Alzheimer’s disease cases: mutation spectrum and cerebrospinal fluid biomarkers. J Alzheimers Dis 2012;30:847–56. [58] Brickell KL, Steinbart EJ, Rumbaugh M, Payami H, Schellenberg GD, Van Deerlin V, et al. Early-onset Alzheimer disease in families with late-onset Alzheimer disease: a potential important subtype of familial Alzheimer disease. Arch Neurol 2006; 63:1307–11.

13

[59] Janssen JC, Beck JA, Campbell TA, Dickinson A, Fox NC, Harvey RJ, et al. Early onset familial Alzheimer’s disease: Mutation frequency in 31 families. Neurology 2003;60:235–9. [60] Dermaut B, Kumar-Singh S, Engelborghs S, Theuns J, Rademakers R, Saerens J, et al. A novel presenilin 1 mutation associated with Pick’s disease but not beta-amyloid plaques. Ann Neurol 2004;55:617–26. [61] Karlstrom H, Brooks WS, Kwok JB, Broe GA, Kril JJ, McCann H, et al. Variable phenotype of Alzheimer’s disease with spastic paraparesis. J Neurochem 2008;104:573–83. [62] Ehling R, Noskova L, Stranecky V, Hartmannova H, Pristoupilova A, Hodanova K, et al. Cerebellar dysfunction in a family harboring the PSEN1 mutation co-segregating with a cathepsin D variant p.A58V. J Neurol Sci 2013;326:75–82. [63] Pericak-Vance MA, Bebout JL, Gaskell PC Jr, Yamaoka LH, Hung WY, Alberts MJ, et al. Linkage studies in familial Alzheimer disease: evidence for chromosome 19 linkage. Am J Hum Genet 1991;48:1034–50. [64] Saunders AM, Strittmatter WJ, Schmechel D, George-Hyslop PH, Pericak-Vance MA, Joo SH, et al. Association of apolipoprotein E allele epsilon 4 with late-onset familial and sporadic Alzheimer’s disease. Neurology 1993;43:1467–72. [65] Strittmatter WJ, Saunders AM, Schmechel D, Pericak-Vance M, Enghild J, Salvesen GS, et al. Apolipoprotein E: high-avidity binding to beta-amyloid and increased frequency of type 4 allele in late-onset familial Alzheimer disease. Proc Natl Acad Sci U S A 1993; 90:1977–81. [66] 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. Science 1993;261:921–3. [67] Farrer LA, Cupples LA, Haines JL, Hyman B, Kukull WA, Mayeux R, et al. Effects of age, sex, and ethnicity on the association between apolipoprotein E genotype and Alzheimer disease. A metaanalysis. APOE and Alzheimer Disease Meta Analysis Consortium. JAMA 1997;278:1349–56. [68] van Duijn CM, Cruts M, Theuns J, Van Gassen G, Backhovens H, Van den Broeck M, et al. Genetic association of the presenilin-1 regulatory region with early- onset Alzheimer’s disease in a population-based sample. Eur J Hum Genet 1999;7:801–6. Q8 [69] Lambert JC, Mann DM, Harris JM, Chartier-Harlin MC, Cumming A, Coates J, et al. The -48 C/T polymorphism in the presenilin 1 promoter is associated with an increased risk of developing Alzheimer’s disease and an increased Abeta load in brain. J Med Genet 2001;38:353–5. [70] Pastor P, Roe CM, Villegas A, Bedoya G, Chakraverty S, Garcia G, et al. Apolipoprotein Eepsilon4 modifies Alzheimer’s disease onset in an E280A PS1 kindred. Ann Neurol 2003;54:163–9. [71] Wijsman EM, Daw EW, Yu X, Steinbart EJ, Nochlin D, Bird TD, et al. APOE and other loci affect age-at-onset in Alzheimer’s disease families with PS2 mutation. Am J Med Genet B Neuropsychiatr Genet 2005;132B:14–20. [72] Corder EH, Saunders AM, Risch NJ, Strittmatter WJ, Schmechel DE, Gaskell PC Jr, et al. Protective effect of apolipoprotein E type 2 allele for late onset Alzheimer disease. Nat Genet 1994;7:180–4. [73] Velez JI, Lopera F, Sepulveda-Falla D, Patel HR, Johar AS, Chuah A, et al. APOE*E2 allele delays age of onset in PSEN1 E280A Alzheimer’s disease. Mol Psychiatry 2015; http://dx.doi.org/10.1038/ mp.2015.177. Q9 [74] Lalli MA, Bettcher BM, Arcila ML, Garcia G, Guzman C, Madrigal L, et al. Whole-genome sequencing suggests a chemokine gene cluster that modifies age at onset in familial Alzheimer’s disease. Mol Psychiatry 2015;20:1294–300. [75] Dermaut B, Croes EA, Rademakers R, Van den Broeck M, Cruts M, Hofman A, et al. PRNP Val129 homozygosity increases risk for early-onset Alzheimer’s disease. Ann Neurol 2003;53:409–12.

REV 5.4.0 DTD  JALZ2140_proof  23 March 2016  6:56 pm  ce

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 1717

14 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

R. Cacace et al. / Alzheimer’s & Dementia - (2016) 1-16

[76] Croes EA, Theuns J, Houwing-Duistermaat JJ, Dermaut B, Sleegers K, Roks G, et al. Octapeptide repeat insertions in the prion protein gene and early onset dementia. J Neurol Neurosurg Psychiatry 2004;75:1166–70. [77] Nicolas G, Charbonnier C, Wallon D, Quenez O, Bellenguez C, Grenier-Boley B, et al. SORL1 rare variants: a major risk factor for familial early-onset Alzheimer’s disease. Mol Psychiatry 2015; http:// dx.doi.org/10.1038/mp.2015.121. [78] Genin E, Hannequin D, Wallon D, Sleegers K, Hiltunen M, Combarros O, et al. APOE and Alzheimer disease: a major gene with semi-dominant inheritance. Mol Psychiatry 2011;16:903–7. [79] Sorbi S, Nacmias B, Forleo P, Piacentini S, Latorraca S, Amaducci L. Epistatic effect of APP717 mutation and apolipoprotein E genotype in familial Alzheimer’s disease. Ann Neurol 1995;38:124–7. [80] Brouwers N, Sleegers K, Engelborghs S, Maurer-Stroh S, Gijselinck I, van der Zee J, et al. Genetic variability in progranulin contributes to risk for clinically diagnosed Alzheimer disease. Neurology 2008;71:656–64. [81] Kelley BJ, Haidar W, Boeve BF, Baker M, Shiung M, Knopman DS, et al. Alzheimer disease-like phenotype associated with the c.154delA mutation in progranulin. Arch Neurol 2010;67:171–7. [82] Cortini F, Fenoglio C, Guidi I, Venturelli E, Pomati S, Marcone A, et al. Novel exon 1 progranulin gene variant in Alzheimer’s disease. Eur J Neurol 2008;15:1111–7. [83] Rademakers R, Dermaut B, Peeters K, Cruts M, Heutink P, Goate A, et al. Tau (MAPT) mutation Arg406Trp presenting clinically with Alzheimer disease does not share a common founder in Western Europe. Hum Mutat 2003;22:409–11. [84] Lindquist SG, Holm IE, Schwartz M, Law I, Stokholm J, Batbayli M, et al. Alzheimer disease-like clinical phenotype in a family with FTDP-17 caused by a MAPT R406W mutation. Eur J Neurol 2008; 15:377–85. [85] Cacace R, Van Cauwenberghe C, Bettens K, Gijselinck I, van der Zee J, Engelborghs S, et al. C9orf72 G4C2 repeat expansions in Alzheimer’s disease and mild cognitive impairment. Neurobiol Aging 2013;34:1712–7. [86] Kohli MA, John-Williams K, Rajbhandary R, Naj A, Whitehead P, Hamilton K, et al. Repeat expansions in the C9ORF72 gene contribute to Alzheimer’s disease in Caucasians. Neurobiol Aging 2012;34:1519.e5–151912. [87] Harms M, Benitez BA, Cairns N, Cooper B, Cooper P, Mayo K, et al. C9orf72 hexanucleotide repeat expansions in clinical Alzheimer disease. JAMA Neurol 2013;70:736–41. [88] Jayadev S, Nochlin D, Poorkaj P, Steinbart EJ, Mastrianni JA, Montine TJ, et al. Familial prion disease with Alzheimer diseaselike tau pathology and clinical phenotype. Ann Neurol 2011; 69:712–20. [89] Guerreiro R, Bras J, Wojtas A, Rademakers R, Hardy J, GraffRadford N. A nonsense mutation in PRNP associated with clinical Alzheimer’s disease. Neurobiol Aging 2014;35:2656.e13–6. [90] Ryman DC, Acosta-Baena N, Aisen PS, Bird T, Danek A, Fox NC, et al. Symptom onset in autosomal dominant Alzheimer disease: a systematic review and meta-analysis. Neurology 2014;83:253–60. [91] Marchani EE, Bird TD, Steinbart EJ, Rosenthal E, Yu CE, Schellenberg GD, et al. Evidence for three loci modifying age-atonset of Alzheimer’s disease in early-onset PSEN2 families. Am J Med Genet B Neuropsychiatr Genet 2010;153B:1031–41. [92] Hardy JA, Higgins GA. Alzheimer’s disease: the amyloid cascade hypothesis. Science 1992;256:184–5. [93] Dimitrov M, Alattia JR, Lemmin T, Lehal R, Fligier A, Houacine J, et al. Alzheimer’s disease mutations in APP but not gamma-secretase modulators affect epsilon-cleavage-dependent AICD production. Nat Commun 2013;4:2246. [94] Morris GP, Clark IA, Vissel B. Inconsistencies and controversies surrounding the amyloid hypothesis of Alzheimer’s disease. Acta Neuropathol Commun 2014;2:135.

[95] Chetelat G, La JR, Villain N, Perrotin A, de La Sayette V, Eustache F, et al. Amyloid imaging in cognitively normal individuals, at-risk populations and preclinical Alzheimer’s disease. Neuroimage Clin 2013; 2:356–65. [96] Serrano-Pozo A, Qian J, Monsell SE, Blacker D, Gomez-Isla T, Betensky RA, et al. Mild to moderate Alzheimer dementia with insufficient neuropathological changes. Ann Neurol 2014;75: 597–601. [97] Jack CR Jr, Wiste HJ, Weigand SD, Knopman DS, Lowe V, Vemuri P, et al. Amyloid-first and neurodegeneration-first profiles characterize incident amyloid PET positivity. Neurology 2013;81:1732–40. [98] Caroli A, Prestia A, Galluzzi S, Ferrari C, van der Flier WM, Ossenkoppele R, et al. Mild cognitive impairment with suspected nonamyloid pathology (SNAP): Prediction of progression. Neurology 2015;84:508–15. [99] Chetelat G. Alzheimer disease: Abeta-independent processesrethinking preclinical AD. Nat Rev Neurol 2013;9:123–4. [100] Dermaut B, Kumar-Singh S, Rademakers R, Theuns J, Cruts M, Van Broeckhoven C. Tau is central in the genetic Alzheimerfrontotemporal dementia spectrum. Trends Genet 2005;21:664–72. [101] van der Zee J, Sleegers K, Van Broeckhoven C. Invited article: the Alzheimer disease-frontotemporal lobar degeneration spectrum. Neurology 2008;71:1191–7. [102] Theuns J, Remacle J, Killick R, Corsmit E, Vennekens K, Huylebroeck D, et al. Alzheimer-associated C allele of the promoter polymorphism -22C.T causes a critical neuron-specific decrease of presenilin 1 expression. Hum Mol Genet 2003;12:869–77. [103] Theuns J, Brouwers N, Engelborghs S, Sleegers K, Bogaerts V, Corsmit E, et al. Promoter mutations that increase amyloid precursor-protein expression are associated with Alzheimer disease. Am J Hum Genet 2006;78:936–46. [104] Brouwers N, Sleegers K, Engelborghs S, Bogaerts V, Serneels S, Kamali K, et al. Genetic risk and transcriptional variability of amyloid precursor protein in Alzheimer’s disease. Brain 2006; 129:2984–91. [105] Cruchaga C, Haller G, Chakraverty S, Mayo K, Vallania FL, Mitra RD, et al. Rare variants in APP, PSEN1 and PSEN2 increase risk for AD in late-onset Alzheimer’s disease families. PLoS One 2012;7:e31039. [106] Beck J, Pittman A, Adamson G, Campbell T, Kenny J, Houlden H, et al. Validation of next-generation sequencing technologies in genetic diagnosis of dementia. Neurobiol Aging 2014; 35:261–5. [107] Clarke AJ. Managing the ethical challenges of next-generation sequencing in genomic medicine. Br Med Bull 2014;111:17–30. [108] Guerreiro RJ, Lohmann E, Kinsella E, Bras JM, Luu N, Gurunlian N, et al. Exome sequencing reveals an unexpected genetic cause of disease: NOTCH3 mutation in a Turkish family with Alzheimer’s disease. Neurobiol Aging 2012;33:1008–23. [109] Pottier C, Hannequin D, Coutant S, Rovelet-Lecrux A, Wallon D, Rousseau S, et al. High frequency of potentially pathogenic SORL1 mutations in autosomal dominant early-onset Alzheimer disease. Mol Psychiatry 2012;17:875–9. [110] Scherzer CR, Offe K, Gearing M, Rees HD, Fang G, Heilman CJ, et al. Loss of apolipoprotein E receptor LR11 in Alzheimer disease. Arch Neurol 2004;61:1200–5. [111] Andersen OM, Reiche J, Schmidt V, Gotthardt M, Spoelgen R, Behlke J, et al. Neuronal sorting protein-related receptor sorLA/ LR11 regulates processing of the amyloid precursor protein. Proc Natl Acad Sci U S A 2005;102:13461–6. [112] Caglayan S, Takagi-Niidome S, Liao F, Carlo AS, Schmidt V, Burgert T, et al. Lysosomal sorting of amyloid-beta by the SORLA receptor is impaired by a familial Alzheimer’s disease mutation. Sci Transl Med 2014;6:223ra20. [113] Rogaeva E, Meng Y, Lee JH, Gu Y, Kawarai T, Zou F, et al. The neuronal sortilin-related receptor SORL1 is genetically associated with Alzheimer disease. Nat Genet 2007;39:168–77.

REV 5.4.0 DTD  JALZ2140_proof  23 March 2016  6:56 pm  ce

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

R. Cacace et al. / Alzheimer’s & Dementia - (2016) 1-16 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

[114] 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. [115] Vardarajan BN, Zhang Y, Lee JH, Cheng R, Bohm C, Ghani M, et al. Coding mutations in SORL1 and Alzheimer disease. Ann Neurol 2015;77:215–27. [116] Bettens K, Sleegers K, Van Broeckhoven C. Genetic insights in Alzheimer’s disease. Lancet Neurol 2013;12:92–104. [117] Sillen A, Forsell C, Lilius L, Axelman K, Bjork BF, Onkamo P, et al. Genome scan on Swedish Alzheimer’s disease families. Mol Psychiatry 2006;11:182–6. [118] Sillen A, Andrade J, Lilius L, Forsell C, Axelman K, Odeberg J, et al. Expanded high-resolution genetic study of 109 Swedish families with Alzheimer’s disease. Eur J Hum Genet 2008;16:202–8. [119] Sillen A, Brohede J, Forsell C, Lilius L, Andrade J, Odeberg J, et al. Linkage analysis of autopsy-confirmed familial Alzheimer disease supports an Alzheimer disease locus in 8q24. Dement Geriatr Cogn Disord 2011;31:109–18. [120] Giedraitis V, Hedlund M, Skoglund L, Blom E, Ingvast S, Brundin R, et al. New Alzheimer’s disease locus on chromosome 8. J Med Genet 2006;43:931–5. [121] Rademakers R, Cruts M, Sleegers K, Dermaut B, Theuns J, Aulchenko Y, et al. Linkage and association studies identify a novel locus for Alzheimer disease at 7q36 in a Dutch population-based sample. Am J Hum Genet 2005;77:643–52. [122] Van Broeckhoven C. The future of genetic research on neurodegeneration. Nat Med 2010;16:1215–7. [123] Valdmanis PN, Belzil VV, Lee J, Dion PA, St-Onge J, Hince P, et al. A mutation that creates a pseudoexon in SOD1 causes familial ALS. Ann Hum Genet 2009;73:652–7. [124] Cruts M, Rademakers R, Gijselinck I, van der Zee J, Dermaut B, De Pooter T, et al. Genomic architecture of human 17q21 linked to frontotemporal dementia uncovers a highly homologous family of low-copy repeats in the tau region. Hum Mol Genet 2005; 14:1753–62. [125] Rademakers R, Melquist S, Cruts M, Theuns J, Del-Favero J, Poorkaj P, et al. High-density SNP haplotyping suggests altered regulation of tau gene expression in progressive supranuclear palsy. Hum Mol Genet 2005;14:3281–92. [126] Cirulli ET, Goldstein DB. Uncovering the roles of rare variants in common disease through whole-genome sequencing. Nat Rev Genet 2010;11:415–25. [127] Nho K, Corneveaux JJ, Kim S, Lin H, Risacher SL, Shen L, et al. Whole-exome sequencing and imaging genetics identify functional variants for rate of change in hippocampal volume in mild cognitive impairment. Mol Psychiatry 2013;18:781–7. [128] Clarimon J, Djaldetti R, Lleo A, Guerreiro RJ, Molinuevo JL, PaisanRuiz C, et al. Whole genome analysis in a consanguineous family with early onset Alzheimer’s disease. Neurobiol Aging 2009; 30:1986–91. [129] Ghani M, Sato C, Lee JH, Reitz C, Moreno D, Mayeux R, et al. Evidence of recessive Alzheimer disease loci in a Caribbean Hispanic data set: genome-wide survey of runs of homozygosity. JAMA Neurol 2013;70:1261–7. [130] Nalls MA, Guerreiro RJ, Simon-Sanchez J, Bras JT, Traynor BJ, Gibbs JR, et al. Extended tracts of homozygosity identify novel candidate genes associated with late-onset Alzheimer’s disease. Neurogenetics 2009;10:183–90. [131] Farrer LA, Bowirrat A, Friedland RP, Waraska K, Korczyn AD, Baldwin CT. Identification of multiple loci for Alzheimer disease in a consanguineous Israeli-Arab community. Hum Mol Genet 2003;12:415–22. [132] Beck JA, Poulter M, Campbell TA, Uphill JB, Adamson G, Geddes JF, et al. Somatic and germline mosaicism in sporadic early-onset Alzheimer’s disease. Hum Mol Genet 2004;13:1219–24.

15

[133] Frigerio CS, Lau P, Troakes C, Deramecourt V, Gele P, Van Loo P, et al. On the identification of low allele frequency mosaic mutations in the brains of Alzheimer disease patients. Alzheimers Dement 2015;11:1265–76. [134] Lardenoije R, Iatrou A, Kenis G, Kompotis K, Steinbusch HW, Mastroeni D, et al. The epigenetics of aging and neurodegeneration. Prog Neurobiol 2015;131:21–64. [135] van den Hove DL, Kompotis K, Lardenoije R, Kenis G, Mill J, Steinbusch HW, et al. Epigenetically regulated microRNAs in Alzheimer’s disease. Neurobiol Aging 2014;35:731–45. [136] De Jager PL, Srivastava G, Lunnon K, Burgess J, Schalkwyk LC, Yu L, et al. Alzheimer’s disease: early alterations in brain DNA methylation at ANK1, BIN1, RHBDF2 and other loci. Nat Neurosci 2014;17:1156–63. [137] Lunnon K, Smith R, Hannon E, De Jager PL, Srivastava G, Volta M, et al. Methylomic profiling implicates cortical deregulation of ANK1 in Alzheimer’s disease. Nat Neurosci 2014;17:1164–70. [138] Davies MN, Volta M, Pidsley R, Lunnon K, Dixit A, Lovestone S, et al. Functional annotation of the human brain methylome identifies tissue-specific epigenetic variation across brain and blood. Genome Biol 2012;13:R43. [139] Skipper M, Eccleston A, Gray N, Heemels T, Le Bot N, Marte B, et al. Presenting the epigenome roadmap. Nature 2015;518:313. [140] Gjoneska E, Pfenning AR, Mathys H, Quon G, Kundaje A, Tsai LH, et al. Conserved epigenomic signals in mice and humans reveal immune basis of Alzheimer’s disease. Nature 2015; 518:365–9. [141] Bennett DA, Yu L, Yang J, Srivastava GP, Aubin C, De Jager PL. Epigenomics of Alzheimer’s disease. Transl Res 2015;165:200–20. [142] Bateman RJ, Xiong C, Benzinger TL, Fagan AM, Goate A, Fox NC, et al. Clinical and biomarker changes in dominantly inherited Alzheimer’s disease. N Engl J Med 2012;367:795–804. [143] Yang Y, Muzny DM, Reid JG, Bainbridge MN, Willis A, Ward PA, et al. Clinical whole-exome sequencing for the diagnosis of mendelian disorders. N Engl J Med 2013;369:1502–11. [144] Fagan AM, Xiong C, Jasielec MS, Bateman RJ, Goate AM, Benzinger TL, et al. Longitudinal change in CSF biomarkers in autosomal-dominant Alzheimer’s disease. Sci Transl Med 2014; 6:226ra30. [145] Reitz C, Brayne C, Mayeux R. Epidemiology of Alzheimer disease. Nat Rev Neurol 2011;7:137–52. [146] Wattamwar PR, Mathuranath PS. An overview of biomarkers in Alzheimer’s disease. Ann Indian Acad Neurol 2010;13:S116–23. [147] Sutphen CL, Jasielec MS, Shah AR, Macy EM, Xiong C, Vlassenko AG, et al. Longitudinal Cerebrospinal Fluid Biomarker Changes in Preclinical Alzheimer Disease During Middle Age. JAMA Neurol 2015;72:1029–42. [148] Toledo JB, Weiner MW, Wolk DA, Da X, Chen K, Arnold SE, et al. Neuronal injury biomarkers and prognosis in ADNI subjects with normal cognition. Acta Neuropathol Commun 2014;2:26. [149] Villemagne VL, Fodero-Tavoletti MT, Masters CL, Rowe CC. Tau imaging: early progress and future directions. Lancet Neurol 2015; 14:114–24. [150] Ayutyanont N, Langbaum JB, Hendrix SB, Chen K, Fleisher AS, Friesenhahn M, et al. The Alzheimer’s prevention initiative composite cognitive test score: sample size estimates for the evaluation of preclinical Alzheimer’s disease treatments in presenilin 1 E280A mutation carriers. J Clin Psychiatry 2014;75:652–60. [151] Schneider LS, Mangialasche F, Andreasen N, Feldman H, Giacobini E, Jones R, et al. Clinical trials and late-stage drug development for Alzheimer’s disease: an appraisal from 1984 to 2014. J Intern Med 2014;275:251–83. [152] Harold D, Abraham R, Hollingworth P, Sims R, Gerrish A, Hamshere ML, et al. Genome-wide association study identifies variants at CLU and PICALM associated with Alzheimer’s disease. Nat Genet 2009;41:1088–93.

REV 5.4.0 DTD  JALZ2140_proof  23 March 2016  6:56 pm  ce

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

16 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

R. Cacace et al. / Alzheimer’s & Dementia - (2016) 1-16

[153] Hollingworth P, Harold D, Sims R, Gerrish A, Lambert JC, Carrasquillo MM, et al. Common variants at ABCA7, MS4A6A/ MS4A4E, EPHA1, CD33 and CD2AP are associated with Alzheimer’s disease. Nat Genet 2011;43:429–35. [154] Lambert JC, 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. [155] Naj AC, Jun G, Beecham GW, Wang LS, Vardarajan BN, Buros J, et al. Common variants at MS4A4/MS4A6E, CD2AP, CD33 and EPHA1 are associated with late-onset Alzheimer’s disease. Nat Genet 2011;43:436–41. [156] Seshadri S, Fitzpatrick AL, Ikram MA, DeStefano AL, Gudnason V, Boada M, et al. Genome-wide analysis of genetic loci associated with Alzheimer disease. JAMA 2010;303:1832–40. [157] Karch CM, Goate AM. Alzheimer’s disease risk genes and mechanisms of disease pathogenesis. Biol Psychiatry 2015;77:43–51. [158] Morgan K. The three new pathways leading to Alzheimer’s disease. Neuropathol Appl Neurobiol 2011;37:353–7. [159] 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.

[160] Jonsson T, Stefansson H, Steinberg S, Jonsdottir I, Jonsson PV, Snaedal J, et al. Variant of TREM2 associated with the risk of Alzheimer’s disease. N Engl J Med 2013;368:107–16. [161] Steinberg S, Stefansson H, Jonsson T, Johannsdottir H, Ingason A, Helgason H, et al. Loss-of-function variants in ABCA7 confer risk of Alzheimer’s disease. Nat Genet 2015;47:445–7. [162] Cuyvers E, De Roeck A, Van den Bossche T, Van Cauwenberghe C, Bettens K, Vermeulen S, et al. Mutations in ABCA7 in a Belgian cohort of Alzheimer’s disease patients: a targeted resequencing study. Lancet Neurol 2015;14:814–22. [163] Korvatska O, Leverenz JB, Jayadev S, McMillan P, Kurtz I, Guo X, et al. R47H Variant of TREM2 Associated With Alzheimer Disease in a Large Late-Onset Family: Clinical, Genetic, and Neuropathological Study. JAMA Neurol 2015;72:920–7. [164] Vardarajan BN, Ghani M, Kahn A, Sheikh S, Sato C, Barral S, et al. Rare coding mutations identified by sequencing of Alzheimer disease genome-wide association studies loci. Ann Neurol 2015; 78:487–98. [165] Guerreiro RJ, Hardy J. Alzheimer’s disease genetics: lessons to improve disease modelling. Biochem Soc Trans 2011;39: 910–6.

REV 5.4.0 DTD  JALZ2140_proof  23 March 2016  6:56 pm  ce

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 2119