This article was downloaded by: [Memorial University of Newfoundland] On: 30 January 2015, At: 14:59 Publisher: Taylor & Francis Informa Ltd Registered in England and Wales Registered Number: 1072954 Registered office: Mortimer House, 37-41 Mortimer Street, London W1T 3JH, UK

Worm Publication details, including instructions for authors and subscription information: http://www.tandfonline.com/loi/kwrm20

Deciphering genetic interactions between ALS genes using C. elegans Martine Therrien a

ab

& J Alex Parker

abc

CRCHUM; Montréal, QC Canada

b

Departement de pathologie et biologie cellulaire; Universite de Montréal; Montréal, QC Canada c

Departement de neurosciences; Universite de Montréal; Montréal, QC Canada Published online: 08 May 2014.

Click for updates To cite this article: Martine Therrien & J Alex Parker (2014) Deciphering genetic interactions between ALS genes using C. elegans, Worm, 3:2, e29047, DOI: 10.4161/worm.29047 To link to this article: http://dx.doi.org/10.4161/worm.29047

PLEASE SCROLL DOWN FOR ARTICLE Taylor & Francis makes every effort to ensure the accuracy of all the information (the “Content”) contained in the publications on our platform. However, Taylor & Francis, our agents, and our licensors make no representations or warranties whatsoever as to the accuracy, completeness, or suitability for any purpose of the Content. Any opinions and views expressed in this publication are the opinions and views of the authors, and are not the views of or endorsed by Taylor & Francis. The accuracy of the Content should not be relied upon and should be independently verified with primary sources of information. Taylor and Francis shall not be liable for any losses, actions, claims, proceedings, demands, costs, expenses, damages, and other liabilities whatsoever or howsoever caused arising directly or indirectly in connection with, in relation to or arising out of the use of the Content. This article may be used for research, teaching, and private study purposes. Any substantial or systematic reproduction, redistribution, reselling, loan, sub-licensing, systematic supply, or distribution in any form to anyone is expressly forbidden. Terms & Conditions of access and use can be found at http:// www.tandfonline.com/page/terms-and-conditions

Commentary

Commentary

Worm 3, e29047; April 2014; © 2014 Landes Bioscience

Deciphering genetic interactions between ALS genes using C. elegans Martine Therrien1,2 and J Alex Parker1,2,3* CRCHUM; Montréal, QC Canada; 2Departement de pathologie et biologie cellulaire; Universite de Montréal; Montréal, QC Canada; Departement de neurosciences; Universite de Montréal; Montréal, QC Canada

1

Downloaded by [Memorial University of Newfoundland] at 14:59 30 January 2015

3

A

Keywords: ALS, C9ORF72, FUS, PGRN, SOD1, TDP-43, neurodegeneration *Correspondence to: J Alex Parker; Email: [email protected] Submitted: 03/05/2014 Revised: 04/14/2014 Accepted: 04/28/2014 http://dx.doi.org/10.4161/worm.29047

myotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder causing selective death of motor neurons in which it is speculated that 10% of cases have a familial history. In the past 20 years, many genes causative for ALS have been discovered, but the link between them and their roles in neurodegeneration remain unknown. The identification of genes associated with both ALS and frontotemporal dementia (FTD), along with the observation of patients affected by both diseases, have suggested that they are part of the same neurodegenerative spectrum. Investigating possible genetic interactions among ALS/FTD genes could help understand the role of these genes in neurodegeneration. To pursue this goal, our group has developed several ALS models to study potential genetic interactions. More recently, we characterized the deletion mutant alfa-1, the ortholog of C9ORF72, to evaluate the potential genetic interactions between C9ORF72/ alfa-1 and other ALS genes. Here, we discuss the genetic interactions identified in our models and how some of these proteins may also be linked to other neurodegenerative disorders. Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder causing selective motor neuron loss. The disease usually starts in the lower limbs, spreading upwards causing death within 3 to 5 y after the onset of symptoms.1,2 Most cases of ALS are sporadic, but it is speculated that 10% are familial.3 Familial and sporadic ALS cases are clinically indistinguishable, thus identifying disease pathogenesis in familial cases would help understand the pathogenesis of sporadic ALS cases. High-throughput

sequencing technologies have been used to great success over the past few years to identify genetic components of ALS. This has led to the rapid identification of more than 20 genes linked to ALS.3 Among the major ALS genes are Superoxide Dismutase 1 (SOD1),4 TAR DNA Binding Protein (TARDBP),5,6 Fused in Sarcoma (FUS),7 and C9ORF72.8,9 The identification of TDP-43, SOD1, OPTN, FUS, UBQLN2, and NEFH in most aggregates seen in ALS patient tissues suggests that these proteins are involved in disease pathogenesis.10 Many hypotheses11-13 have been put forth regarding the different toxic pathways leading to neurodegeneration; however, no consensus has been reached. Perhaps, more importantly, there are no drugs that effectively slow or halt neuronal loss in ALS patients. An unfortunate recurring theme is that many drugs that were promising candidates in preclinical trials using rodent models have failed in humans.14 Many of these compounds focused on mechanisms to reduce protein aggregation or neuronal death, but recent hypotheses suggest that drugs targeting early pathogenic events could be more effective. However, since ALS diagnosis is difficult and disease progression is rapid, the identification of biological markers is essential for effective therapeutic intervention.

Genetic Interaction Among ALS Genes Many genes linked to ALS appear to have common cellular function. For example, RNA metabolism is a recurrent theme with TDP-43, FUS,

www.landesbioscience.com Worm e29047-1

Downloaded by [Memorial University of Newfoundland] at 14:59 30 January 2015

Figure  1. An emerging genetic network for neurodegeneration. A genetic network of genes underlying neurodegeneration demonstrating the central role of genes linked to the ALS-FTD disease spectrum. Tractable genetic systems like C. elegans are essential to investigate this genetic network of neuronal degeneration. Listed are human proteins linked to disease along with their C. elegans ortholog in parentheses. Abbreviations: ALS, amyotrophic lateral sclerosis; FTD, frontotemporal dementia; FUS, fused in sarcoma; MAPT, microtubule associated protein tau; PGRN, progranulin; SOD1, superoxide dismutase 1; UBQLN2, ubiquilin 2.

HNRNPA2B1, SETX, HNRNPA1, and ARHGEF28 all being involved in this pathway.3 A better understanding of the normal biological roles and the specific pathways affected by mutations may shed light on pathogenic mechanisms in the disease state. Work toward this goal has been done to identify protein–protein interaction networks and protein–RNA interactions of ALS proteins. However, proteins like TDP-43 and FUS have numerous roles affecting the metabolism of thousands of RNAs.12 Also, cells have many compensating pathways, so the genes involved in ALS might not all function in the same pathway, or interact directly but rather be in pathways that have similar functions. Thus, a genetic network of ALS genes would be an essential tool to the understanding of ALS pathogenesis. Finding the relation between ALS genes could be done through the analysis of possible genetic interactions. Genetic interactions are not always easy to determine, but if genes interact it may mean they function in the same pathway or in a compensating cellular pathway.15 To study genetic pathways, model organisms are essential. Analyzing genetic interactions requires models that can

be easily genetically modified and in which phenotypes can be quickly and easily observed. An increasingly popular model with these characteristics is the nematode Caenorhabditis elegans. Our laboratory aims, with the use of different C. elegans models, to unravel some of the interactions in this putative ALS genetic network. To do so, we have developed several C. elegans models to study ALS. The expression of the mutant TDP-43A315T or FUSS57Δ proteins in the animal’s GABAergic motor neurons results in age-dependent motility defects, neurodegeneration, and increased endoplasmic reticulum (ER) stress,16,17 recapitulating important features of ALS pathogenesis. The loss-of-function mutant of tdp1, the C. elegans ortholog of TDP-43, decrease the toxicity observed in these transgenic animals, suggesting an interaction between TDP-1 and TDP-43 and TDP-1 and FUS.17 Unpublished results using fust-1, the worm ortholog of FUS, deletion mutant, also suggest that it participates in mutant TDP-43 and mutant FUS proteotoxicity. Using these models we have determined that TDP-1/TDP43 and FUST-1/FUS genetically interact and modify the progression of motor neuron degeneration. In a recent publication, we aimed to model the toxicity of C9ORF72. In patients, a pathogenic GGGGCC repeat was identified in the first intron of C9ORF72.8,18 The function of C9ORF72 is not fully known but a recent study suggests it has roles in endosomal trafficking.19 However, it is still unclear if the mutation results in a loss of function, a gain of function, or both.20 In patients, hypermethylation of the pathoghenic GGGGCC repeat was shown to decrease the expression of C9ORF72.18,21,22 To model the decreased expression of C9ORF72, we examined the C. elegans ortholog alfa-1 (ALS/FTD associated gene homolog) and a mutation harboring a deletion of the third and fourth exons of the gene leading to a drastic reduction in gene expression. Mutant alfa-1 worms exhibited motility defects leading to an age-dependent paralysis and the degeneration of GABAergic motor neurons.23 Mutant worms also showed stress

e29047-2 Worm

sensitivity, with osmotic stress provoking motor neuron degeneration.23 The pathogenic GGGGCC repeat found in C9ORF72 is the most significant cause of familial and sporadic ALS identified thus far.3 We do not yet know the function of C9ORF72 and if it is functionally similar to other ALS genes. Importantly, for many ALS cases negative for the pathogenic GGGGCC repeat, the expression level of C9ORF72 was also reduced,24 suggesting that C9ORF72 is an important player in ALS pathogenesis. To further our understanding of C9ORF72 pathogenesis, we generated worms expressing mutant TDP-43A315T or FUSS57Δ proteins in combination with the null mutation for alfa-1. We observed that decreased expression of alfa-1 exacerbated the paralysis phenotype in worms expressing mutant TDP-43 but did not alter the toxicity of mutant FUS proteins. These results suggest that C9ORF72 functions in the same toxic pathway as FUS, and perhaps independently (or in parallel) from TDP-43. Therefore, even though FUS and TDP-43 share many structural similarities, they interact differently with alfa-1/C9ORF72 in motor neuron toxicity.

ALS Genes Link to Other Neurodegenerative Disorders? Many genes linked to ALS are also implicated in other neurodegenerative disorders. For example, many ALS proteins were found in aggregates of other neurodegenerative disorders.25-29 It is still unknown if the recruitment of proteins to aggregates is a specific pathological event or not 30 and how it could causes toxicity. It has been speculated that the recruitment of these proteins to aggregates may produce a partial loss of function at the cellular level, but many questions remain about this mechanism in regards to neuronal function and neurodegeneration. Frontotemporal dementia (FTD) also seems to be connected to many other neurodegenerative disorders. The identification of genetic mutations in C9ORF72, TARDBP, and UBQLN2 in FTD and ALS,5,8,18,31 has led to the acceptance of a continuum between these

Volume 3

Downloaded by [Memorial University of Newfoundland] at 14:59 30 January 2015

diseases. Furthermore, genetic mutations in MAPT, PGRN, and C9ORF72 are also the main causes of familial FTD.32 Mutations in MAPT have also been linked to a susceptibility loci of Parkinson disease33-35 and is a major player in Alzheimer disease.36 Therefore, FTD appears to network with many other neurodegenerative disorders and shares many toxic mechanisms with ALS. We think the genetic network including ALS can also incorporate polyglutamine diseases, Alzheimer, and Parkinson diseases. FUS and TDP-43 were shown to colocalize to aggregates in models of the polyglutamine expansion disorders including, Huntington’s Disease and Machado-Joseph Disease,37,38 and to interact with ATXN-239,40 the protein linked to spinocerebellar ataxia type 2. Moreover, in worms, wild-type TDP-1 and FUST-1 proteins contribute the agedependent toxicity of mutant polyglutamine proteins.41 Finally, ER stress has been a recurrent theme regarding ALS pathogenesis in many models as well as in patients.13,42 Similar toxic mechanisms were also implicated in other neurodegenerative disorders, including Parkinson disease and Alzheimer disease.43,44

Concluding Remarks In the motor neuron disease hereditary spastic paraplegia (HSP), characterization of a network including many HSP genes has proven successful in identifying new genes and novel neurodegeneration pathways.45 Furthermore, the network identified proved to be highly related to Parkinson disease, Alzheimer disease, and ALS, further suggesting a link between neurodegenerative diseases. We believe the development of a similar network regarding ALS is essential for our understanding of the disease state and for the identification of biological markers. With the recent identification of the non-coding GGGGCC repeat found in the first intron of C9ORF72 link with ALS and FTD, we continued our investigation of a genetic network among ALS genes. We have shown that a decreased expression of alfa-1/C9ORF72 enhanced the toxicity of mutant TDP-43 proteins

but not FUS.23 We suggest a genetic network (Fig. 1) that includes many genes involved in ALS and FTD where FUS and TDP-43 interact with each other but have distinct effect on C9ORF72 and PGRN17,41 Of course, much more remains to be learned about C9ORF72 biology and pathology. We still do not know why individuals will develop ALS or FTD or both. The close relationship between FTD and other neurodegenerative disorders, such as Parkinson disease and Alzheimer disease, suggest that the characterization of C9ORF72 toxicity is essential and will uncover important pathogenic mechanisms related to many neurodegenerative diseases. The involvement of C9ORF72/ ALFA-1 in the osmotic stress response and the induction of neuronal breaks by an osmotic stress are particularly interesting in this regard. The osmotic stress response is important to maintain proper cell size and protein homeostasis.46 Stress response and protein misfolding are two important aspects of ALS pathogenesis as well as in other neurodegenerative disorders. The development of a genetic network could help understand the importance of those toxic mechanisms in the neuronal loss and of the role of the participating proteins in this pathway. Disclosure of Potential Conflicts

No potential conflicts of interest were disclosed. References 1. Robberecht W, Philips T. The changing scene of amyotrophic lateral sclerosis. Nat Rev Neurosci 2013; 14:248-64; PMID:23463272; http://dx.doi. org/10.1038/nrn3430 2. Kiernan MC. ALS and neuromuscular disease: in search of the Holy Grail. Lancet Neurol 2014; 13:134; PMID:24331786; http://dx.doi.org/10.1016/ S1474-4422(13)70226-6 3. Renton AE, Chiò A, Traynor BJ. State of play in amyotrophic lateral sclerosis genetics. Nat Neurosci 2014; 17:17-23; PMID:24369373; http://dx.doi. org/10.1038/nn.3584 4. Rosen DR, Siddique T, Patterson D, Figlewicz DA, Sapp P, Hentati A, Donaldson D, Goto J, O’Regan JP, Deng HX, et al. Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature 1993; 362:59-62; PMID:8446170; http://dx.doi.org/10.1038/362059a0 5. Kabashi E, Valdmanis PN, Dion P, Spiegelman D, McConkey BJ, Vande Velde C, Bouchard J-P, Lacomblez L, Pochigaeva K, Salachas F, et al. TARDBP mutations in individuals with sporadic and familial amyotrophic lateral sclerosis. Nat Genet 2008; 40:572-4; PMID:18372902; http://dx.doi. org/10.1038/ng.132

6. Sreedharan J, Blair IP, Tripathi VB, Hu X, Vance C, Rogelj B, Ackerley S, Durnall JC, Williams KL, Buratti E, et al. TDP-43 mutations in familial and sporadic amyotrophic lateral sclerosis. Science 2008; 319:1668-72; PMID:18309045; http://dx.doi. org/10.1126/science.1154584 7. Vance C, Rogelj B, Hortobágyi T, De Vos KJ, Nishimura AL, Sreedharan J, Hu X, Smith B, Ruddy D, Wright P, et al. Mutations in FUS, an RNA processing protein, cause familial amyotrophic lateral sclerosis type 6. Science 2009; 323:1208-11; PMID:19251628; http://dx.doi.org/10.1126/science.1165942 8. Renton AE, Majounie E, Waite A, Simón-Sánchez J, Rollinson S, Gibbs JR, Schymick JC, Laaksovirta H, van Swieten JC, Myllykangas L, et al.; ITALSGEN Consortium. A hexanucleotide repeat expansion in C9ORF72 is the cause of chromosome 9p21-linked ALS-FTD. Neuron 2011; 72:257-68; PMID:21944779; http://dx.doi.org/10.1016/j. neuron.2011.09.010 9. Del Villar K, Miller CA. Down-regulation of DENN/ MADD, a TNF receptor binding protein, correlates with neuronal cell death in Alzheimer’s disease brain and hippocampal neurons. Proc Natl Acad Sci U S A 2004; 101:4210-5; PMID:15007167; http://dx.doi. org/10.1073/pnas.0307349101 10. Al-Chalabi A, Jones A, Troakes C, King A, Al-Sarraj S, van den Berg LH. The genetics and neuropathology of amyotrophic lateral sclerosis. Acta Neuropathol 2012; 124:339-52; PMID:22903397; http://dx.doi. org/10.1007/s00401-012-1022-4 11. Pickles S, Vande Velde C. Misfolded SOD1 and ALS: zeroing in on mitochondria. Amyotroph Lateral Scler 2012; 13:333-40; PMID:22471903; http://dx.doi.org /10.3109/17482968.2012.648645 12. Lagier-Tourenne C, Polymenidou M, Hutt KR, Vu AQ, Baughn M, Huelga SC, Clutario KM, Ling S-C, Liang TY, Mazur C, et al. Divergent roles of ALS-linked proteins FUS/TLS and TDP-43 intersect in processing long pre-mRNAs. Nat Neurosci 2012; 15:1488-97; PMID:23023293; http://dx.doi.org/10.1038/nn.3230 13. Matus S, Valenzuela V, Medinas DB, Hetz C. ER Dysfunction and Protein Folding Stress in ALS. Int J Cell Biol 2013; 2013:674751; PMID:24324498; http://dx.doi.org/10.1155/2013/674751 14. Perrin S. Preclinical research: Make mouse studies work. Nature 2014; 507:423-5; PMID:24678540; http://dx.doi.org/10.1038/507423a 15. Boucher B, Jenna S. Genetic interaction networks: better understand to better predict. Front Genet 2013; 4:290; PMID:24381582; http://dx.doi.org/10.3389/ fgene.2013.00290 16. Vaccaro A, Tauffenberger A, Aggad D, Rouleau G, Drapeau P, Parker JA. Mutant TDP-43 and FUS cause age-dependent paralysis and neurodegeneration in C. elegans. PLoS One 2012; 7:e31321; PMID:22363618; http://dx.doi.org/10.1371/journal.pone.0031321 17. Vaccaro A, Tauffenberger A, Ash PEA, Carlomagno Y, Petrucelli L, Parker JA. TDP-1/TDP-43 regulates stress signaling and age-dependent proteotoxicity in Caenorhabditis elegans. PLoS Genet 2012; 8:e1002806; PMID:22792076; http://dx.doi. org/10.1371/journal.pgen.1002806 18. DeJesus-Hernandez M, Mackenzie IR, Boeve BF, Boxer AL, Baker M, Rutherford NJ, Nicholson AM, Finch NA, Flynn H, Adamson J, et al. Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron 2011; 72:245-56; PMID:21944778; http://dx.doi.org/10.1016/j.neuron.2011.09.011 19. Farg MA, Sundaramoorthy V, Sultana JM, Yang S, Atkinson RAK, Levina V, Halloran MA, Gleeson PA, Blair IP, Soo KY, et al. C9ORF72, implicated in amytrophic lateral sclerosis and frontotemporal dementia, regulates endosomal trafficking. Hum Mol Genet 2014; (Forthcoming); PMID:24549040

www.landesbioscience.com Worm e29047-3

Downloaded by [Memorial University of Newfoundland] at 14:59 30 January 2015

20. Ling S-C, Polymenidou M, Cleveland DW. Converging mechanisms in ALS and FTD: disrupted RNA and protein homeostasis. Neuron 2013; 79:41638; PMID:23931993; http://dx.doi.org/10.1016/j. neuron.2013.07.033 21. Xi Z, Zinman L, Moreno D, Schymick J, Liang Y, Sato C, Zheng Y, Ghani M, Dib S, Keith J, et al. Hypermethylation of the CpG island near the G4C2 repeat in ALS with a C9orf72 expansion. Am J Hum Genet 2013; 92:981-9; PMID:23731538; http:// dx.doi.org/10.1016/j.ajhg.2013.04.017 22. Belzil VV, Bauer PO, Prudencio M, Gendron TF, Stetler CT, Yan IK, Pregent L, Daughrity L, Baker MC, Rademakers R, et al. Reduced C9orf72 gene expression in c9FTD/ALS is caused by histone trimethylation, an epigenetic event detectable in blood. Acta Neuropathol 2013; 126:895-905; PMID:24166615; http://dx.doi. org/10.1007/s00401-013-1199-1 23. Therrien M, Rouleau GA, Dion PA, Parker JA. Deletion of C9ORF72 results in motor neuron degeneration and stress sensitivity in C. elegans. PLoS One 2013; 8:e83450; PMID:24349511; http://dx.doi. org/10.1371/journal.pone.0083450 24. Ciura S, Lattante S, Le Ber I, Latouche M, Tostivint H, Brice A, Kabashi E. Loss of function of C9orf72 causes motor deficits in a zebrafish model of Amyotrophic Lateral Sclerosis. Ann Neurol 2013; (Forthcoming); PMID:23720273 25. Neumann M, Sampathu DM, Kwong LK, Truax AC, Micsenyi MC, Chou TT, Bruce J, Schuck T, Grossman M, Clark CM, et al. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science 2006; 314:130-3; PMID:17023659; http://dx.doi.org/10.1126/science.1134108 26. Mackenzie IR, Rademakers R, Neumann M. TDP-43 and FUS in amyotrophic lateral sclerosis and frontotemporal dementia. Lancet Neurol 2010; 9:9951007; PMID:20864052; http://dx.doi.org/10.1016/ S1474-4422(10)70195-2 27. Woulfe J, Gray DA, Mackenzie IRA. FUSimmunoreactive intranuclear inclusions in neurodegenerative disease. Brain Pathol 2010; 20:589-97; PMID:19832837; http://dx.doi. org/10.1111/j.1750-3639.2009.00337.x 28. Mori F, Tanji K, Toyoshima Y, Sasaki H, Yoshida M, Kakita A, Takahashi H, Wakabayashi K. Valosincontaining protein immunoreactivity in tauopathies, synucleinopathies, polyglutamine diseases and intranuclear inclusion body disease. Neuropathology 2013; 33:637-44; PMID:23782134; http://dx.doi. org/10.1111/neup.12050 29. Kon T, Mori F, Tanji K, Miki Y, Toyoshima Y, Yoshida M, Sasaki H, Kakita A, Takahashi H, Wakabayashi K. ALS-associated protein FIG4 is localized in Pick and Lewy bodies, and also neuronal nuclear inclusions, in polyglutamine and intranuclear inclusion body diseases. Neuropathology 2014; 34:19-26; PMID:23888880; http://dx.doi.org/10.1111/ neup.12056

30. Wolfe KJ, Cyr DM. Amyloid in neurodegenerative diseases: friend or foe? Semin Cell Dev Biol 2011; 22:47681; PMID:21458579; http://dx.doi.org/10.1016/j. semcdb.2011.03.011 31. Deng H-X, Chen W, Hong S-T, Boycott KM, Gorrie GH, Siddique N, Yang Y, Fecto F, Shi Y, Zhai H, et al. Mutations in UBQLN2 cause dominant X-linked juvenile and adult-onset ALS and ALS/dementia. Nature 2011; 477:211-5; PMID:21857683; http://dx.doi. org/10.1038/nature10353 32. Warren JD, Rohrer JD, Rossor MN. Clinical review. Frontotemporal dementia. BMJ 2013; 347:f4827; PMID:23920254; http://dx.doi.org/10.1136/bmj. f4827 33. Healy DG, Abou-Sleiman PM, Lees AJ, Casas JP, Quinn N, Bhatia K, Hingorani AD, Wood NW. Tau gene and Parkinson’s disease: a case-control study and meta-analysis. J Neurol Neurosurg Psychiatry 2004; 75:962-5; PMID:15201350; http://dx.doi. org/10.1136/jnnp.2003.026203 34. Pankratz N, Wilk JB, Latourelle JC, DeStefano AL, Halter C, Pugh EW, Doheny KF, Gusella JF, Nichols WC, Foroud T, et al.; PSG-PROGENI and GenePD Investigators, Coordinators and Molecular Genetic Laboratories. Genomewide association study for susceptibility genes contributing to familial Parkinson disease. Hum Genet 2009; 124:593605; PMID:18985386; http://dx.doi.org/10.1007/ s00439-008-0582-9 35. Simón-Sánchez J, Schulte C, Bras JM, Sharma M, Gibbs JR, Berg D, Paisan-Ruiz C, Lichtner P, Scholz SW, Hernandez DG, et al. Genome-wide association study reveals genetic risk underlying Parkinson’s disease. Nat Genet 2009; 41:1308-12; PMID:19915575; http://dx.doi.org/10.1038/ng.487 36. Mohamed N-V, Herrou T, Plouffe V, Piperno N, Leclerc N. Spreading of tau pathology in Alzheimer’s disease by cell-to-cell transmission. Eur J Neurosci 2013; 37:1939-48; PMID:23773063; http://dx.doi. org/10.1111/ejn.12229 37. Doi H, Okamura K, Bauer PO, Furukawa Y, Shimizu H, Kurosawa M, Machida Y, Miyazaki H, Mitsui K, Kuroiwa Y, et al. RNA-binding protein TLS is a major nuclear aggregate-interacting protein in huntingtin exon 1 with expanded polyglutamine-expressing cells. J Biol Chem 2008; 283:6489-500; PMID:18167354; http://dx.doi.org/10.1074/jbc.M705306200 38. Tan C-F, Yamada M, Toyoshima Y, Yokoseki A, Miki Y, Hoshi Y, Kaneko H, Ikeuchi T, Onodera O, Kakita A, et al. Selective occurrence of TDP-43immunoreactive inclusions in the lower motor neurons in Machado-Joseph disease. Acta Neuropathol 2009; 118:553-60; PMID:19526244; http://dx.doi. org/10.1007/s00401-009-0552-x

e29047-4 Worm

39. Elden AC, Kim H-J, Hart MP, Chen-Plotkin AS, Johnson BS, Fang X, Armakola M, Geser F, Greene R, Lu MM, et al. Ataxin-2 intermediate-length polyglutamine expansions are associated with increased risk for ALS. Nature 2010; 466:1069-75; PMID:20740007; http://dx.doi.org/10.1038/nature09320 40. Farg MA, Soo KY, Warraich ST, Sundaramoorthy V, Blair IP, Atkin JD. Ataxin-2 interacts with FUS and intermediate-length polyglutamine expansions enhance FUS-related pathology in amyotrophic lateral sclerosis. Hum Mol Genet 2013; 22:717-28; PMID:23172909; http://dx.doi.org/10.1093/hmg/ dds479 41. Tauffenberger A, Chitramuthu BP, Bateman A, Bennett HPJ, Parker JA. Reduction of polyglutamine toxicity by TDP-43, FUS and progranulin in Huntington’s disease models. Hum Mol Genet 2013; 22:782-94; PMID:23172908; http://dx.doi. org/10.1093/hmg/dds485 42. Musarò A. Understanding ALS: new therapeutic approaches. FEBS J 2013; 280:4315-22; PMID:23217177; http://dx.doi.org/10.1111/ febs.12087 43. Hoozemans JJM, Scheper W. Endoplasmic reticulum: the unfolded protein response is tangled in neurodegeneration. Int J Biochem Cell Biol 2012; 44:12958; PMID:22564438; http://dx.doi.org/10.1016/j. biocel.2012.04.023 44. Mercado G, Valdés P, Hetz C. An ERcentric view of Parkinson’s disease. Trends Mol Med 2013; 19:16575; PMID:23352769; http://dx.doi.org/10.1016/j. molmed.2012.12.005 45. Novarino G, Fenstermaker AG, Zaki MS, Hofree M, Silhavy JL, Heiberg AD, Abdellateef M, Rosti B, Scott E, Mansour L, et al. Exome sequencing links corticospinal motor neuron disease to common neurodegenerative disorders. Science 2014; 343:50611; PMID:24482476; http://dx.doi.org/10.1126/ science.1247363 46. Lamitina T, Huang CG, Strange K. Genome-wide RNAi screening identifies protein damage as a regulator of osmoprotective gene expression. Proc Natl Acad Sci U S A 2006; 103:12173-8; PMID:16880390; http://dx.doi.org/10.1073/pnas.0602987103

Volume 3

Deciphering genetic interactions between ALS genes using C. elegans.

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder causing selective death of motor neurons in which it is speculated that 10% ...
175KB Sizes 0 Downloads 6 Views