Next-generation sequencing has greatly accelerated the discovery of rare human genetic diseases. Nearly 45% of patients have variants associated with known diseases but the unsolved cases remain a conundrum. Moreover, causative mutations can be difficult to pinpoint because variants frequently map to genes with no previous disease associations and, often, only one or a few patients with variants in the same gene are identified. Model organisms, such as Drosophila, can help to identify and characterize these new disease-causing genes. Importantly, Drosophila allow quick and sophisticated genetic manipulations, permit functional testing of human variants, enable the characterization of pathogenic mechanisms and are amenable to drug tests. In this Spotlight, focusing on microcephaly as a case study, we highlight how studies of human genes in Drosophila have aided our understanding of human genetic disorders, allowing the identification of new genes in well-established signaling pathways.

Every in vivo approach used to study genetic variants associated with human disease has benefits and caveats. Whether the system is based on human cells in culture, primates, rodents, fish, flies, worms or yeast, no model can recapitulate every aspect of human development and disease. In addition, the speed and costs associated with approaches in various model organisms can vary by orders of magnitude. Yet, effectively addressing how genetic variation might lead to human disease is important, both for diagnosis and for developing suitable therapies.

A number of established genetic model systems, such as worms, flies, fish and mice, offer the ability to test specific variants associated with human disorders in vivo (Wangler et al., 2017a). The fruit fly Drosophila melanogaster is particularly well suited for these studies because of the ease and sophistication of its genetic manipulation, its short generation time and the fact that about 75% of human genes have Drosophila homologs (Chow and Reiter, 2017; Reiter et al., 2001). Indeed, research on Drosophila in the Model Organism Screening Center (MOSC) of the Undiagnosed Diseases Network (see below) and in collaboration with other human geneticists has provided diagnoses for more than 25 rare human genetic diseases in less than 4 years (Bellen et al., 2019; Wangler et al., 2017a; 2017b). These include diseases caused by proteins associated with different organelles, such as mitochondria (Chao et al., 2017; Harel et al., 2016; Liu et al., 2017; Oláhová et al., 2018; Yoon et al., 2017), peroxisomes (Chao et al., 2016; Chung et al., 2020; Luo et al., 2017; Wangler et al., 2017b), lysosomes (Şentürk et al., 2019) and endosomes (Lin et al., 2018), as well as numerous developmental disorders (Ansar et al., 2018a; Ansar et al., 2018b; Link et al., 2019; Marcogliese et al., 2018).

Here, we provide an overview of how studies of Drosophila have contributed to the identification and characterization of human diseases. We then discuss the recent discovery of a number of genes associated with microcephaly in humans. These discoveries provide us with a better understanding of the molecular mechanisms underlying asymmetric cell division in neuroblasts in Drosophila, which in turn has led to the discovery of many homologs of Drosophila genes that are associated with microcephaly in humans.

In the past, human geneticists mostly relied on large family pedigrees to identify genes associated with disease (Wijsman, 2012). However, with the advent of next-generation sequencing technology, it is now often possible to identify one or a few variants that are associated with disease in a single individual (Bamshad et al., 2019; Posey et al., 2019). Typically, 40-50% of pediatric diseases can be diagnosed by identifying variants in previously described disease genes using whole-exome sequencing (WES) combined with assessment of copy number variations (CNVs) (Liu et al., 2019; Ngo et al., 2020), or by performing whole-genome sequencing (WGS). However, the remaining cases (∼50%) go undiagnosed (Adams and Eng, 2018). If additional tests or experiments are performed, a significant number of cases can eventually be diagnosed (Ramoni et al., 2017; Splinter et al., 2018). These often include sequencing the DNA of the patient's parents and other family members, the identification of other independent affected individuals through gene-matching algorithms (Buske et al., 2015; Sobreira et al., 2015) and RNA sequencing of blood or fibroblasts (Frésard et al., 2019).

Despite these efforts, there are still millions of individuals with undiagnosed diseases, and it is estimated that 6000-13,000 human genes remain to be associated with diseases (Bamshad et al., 2019). The National Institutes of Health has established and supported consortia such as the Centers for Mendelian Genomics (CMG) to uncover the genetics of Mendelian disease (Bamshad et al., 2012; Posey et al., 2019) and the Undiagnosed Diseases Network (UDN) (Ramoni et al., 2017; Splinter et al., 2018) to diagnose patients with rare diseases who typically undergo a long diagnostic odyssey. Other consortia include Finding of Rare Disease Genes (FORGE) (Beaulieu et al., 2014), Care4Rare Canada Consortium (Sawyer et al., 2016), Canadian Rare Diseases Models and Mechanisms Network (RDMM) (Boycott et al., 2020), Deciphering Developmental Disorders (DDD) (Firth and Wright, 2011), Undiagnosed Diseases Network International (UDNI) (Taruscio et al., 2015) and the International Rare Diseases Research Consortium (IRDiRC) (Gasser et al., 2012).

When a diagnosis cannot be reached for an individual after analyzing clinical and genetic information, genetic model organisms can be used to model human disease variants and assess their function in a developing or adult animal. Drosophila has been very successful in this respect, as 50-70% of human genes can rescue phenotypes associated with loss of the homologous gene in Drosophila (Bellen et al., 2019). Importantly, 75% of human genes have Drosophila orthologs (Reiter et al., 2001). Finally, 65% of the applicants to the UDN are children, and 65-70% have neurological symptoms (Splinter et al., 2018; Wangler et al., 2017a), highlighting that many undiagnosed and rare diseases are developmental in nature, often affecting neuronal development or function.

Pathways that regulate brain development are largely conserved between Drosophila and humans (Hirth and Reichert, 1999; Pires-daSilva and Sommer, 2003), and the Drosophila brain undergoes many of the same developmental processes as the human brain. Neuronal stem cell division and neurogenesis have been extensively investigated using Drosophila neuroblasts and their distinct lineages (Gallaud et al., 2017; Homem and Knoblich, 2012). Furthermore, the Drosophila nervous system has been crucial to the study of processes that regulate axonal guidance (Sáchez-Soriano et al., 2007) and the formation of neural circuits (Davis et al., 2020; Olsen and Wilson, 2008; Zheng et al., 2018). Hence, the developing brain (larval and pupal stages) or the adult brain in Drosophila can be used as models of human brain development and neurodegeneration and are useful to decipher human neurological disease; there are, of course, some aspects of human brain development and morphological features that cannot be effectively modeled in Drosophila. In addition, although ∼75% human genes do have a Drosophila ortholog, there are some (the remaining ∼25%) that do not, so investigations focused on these would be better suited in other model systems. In most cases, genes that cannot be modeled in flies due to lack of homologs can effectively be modeled in fish or mice.

To test the function of a human variant, and whether it might cause disease, Drosophila can be ‘humanized’. This involves expressing the human protein in a Drosophila mutant background to assess whether it can rescue phenotypes associated with gene loss. This is most often achieved by using GAL4 drivers (Brand and Perrimon, 1993; Caygill and Brand, 2016; Duffy, 2002) or by inserting a disruptive GAL4 in the Drosophila gene (Bellen and Yamamoto, 2015; Diao et al., 2015) to drive a UAS-human reference cDNA (Kanca et al., 2017; Şentürk et al., 2019). If the human protein encoded by the cDNA can rescue phenotypes associated with loss-of-function mutations in Drosophila, then the function of the protein is conserved. Human variants can then be tested, and the following questions can be addressed: (1) Does the putative disease variant rescue the mutant phenotype or not; if so, how do the phenotypes associated with variant expression differ from those associated with the reference protein? (2) Does the reference or disease variant protein cause phenotypes in a wild-type animal, suggesting that they are dominant-negative or gain-of-function variants? (3) Can the function of the protein, and hence the mechanisms of disease pathogenesis, be tackled? (4) Do the identified proteins/pathways provide opportunities for development of therapies and drug testing? Answering these questions can provide a diagnosis, link many genes to known pathways and processes, and identify drugs that can be tested in patients (Bellen et al., 2019; Chung et al., 2020; Yoon et al., 2017).

An alternative approach to identifying genes associated with human disease is to carry out a forward genetic screen that probes disease-associated phenotypes in Drosophila. One such screen has been successful at identifying novel genes associated with human disease (Yamamoto et al., 2014), identifying 165 loci associated with developmental or neurodegenerative phenotypes. Of these, 93% had human homologs, and 61% are currently associated with human diseases based on MARRVEL (model organism aggregated resources for rare variant exploration, Wangler et al., 2017a), an online database integrating genetic information from humans and across species to provide a unified interface for gene, protein or disease investigation. However, 30% of the genes that are submitted by the UDN consortium have not been isolated in this or other genetic screens and have almost no annotation in any model organism. Moreover, existing annotations can be restricted to one species and are frequently cursory in nature (Wangler et al., 2017b). Hence, defining the function of human genes and their homologs associated with developmental diseases can provide crucial biological information about conserved developmental processes.

In recent years, several developmental disorders have been identified, and the function of the corresponding genes have been characterized using Drosophila. These include genes associated with autism spectrum disorder (ASD) (reviewed by Bellosta and Soldano, 2019) and neurological syndromes that encompass developmental delay, intellectual disability, seizures or psychiatric problems (Halperin et al., 2019; Hubert et al., 2020; Kummeling et al., 2020; Muir et al., 2020; Nixon et al., 2019; Owings et al., 2018; Straub et al., 2018). Another example is congenital microcephaly (Jayaraman et al., 2018; Schoborg et al., 2019): a developmental disorder that can be caused by genetic mutation, virus exposure or environmental toxins (Devakumar et al., 2018; Pirozzi et al., 2018). Primary microcephaly is defined as a disorder in which individuals exhibit a smaller than normal head size, as measured by an occipital frontal circumference (OFC) that is 2-3 standard deviations (s.d.) below the average for the age and gender of the individual. Currently, 25 genetic loci are associated with primary autosomal recessive microcephaly (MCPH) (Jayaraman et al., 2018; Naveed et al., 2018) in the Online Mendelian Inheritance in Man (OMIM) database (Amberger et al., 2015). In recent years, the molecular function of some of these MCPH genes has been elucidated using Drosophila (Gambarotto et al., 2019; Lucas and Raff, 2007; Poulton et al., 2017; Ramdas Nair et al., 2016; Rujano et al., 2013; Schoborg et al., 2015; Schoborg et al., 2019; Singh et al., 2014). Below, we outline how studies of Drosophila have played an important role in elucidating some of the pathogenic mechanisms associated with microcephaly and in aiding microcephaly disease diagnosis.

Heterozygous missense mutations in the gene ALFY (also termed WDFY3) were recently identified in a large family with autosomal dominant primary microcephaly (Kadir et al., 2016). ALFY encodes a protein that functions in the removal of aggregated proteins by serving as a scaffold for autophagy-mediated removal (Filimonenko et al., 2010; Simonsen et al., 2004). To determine whether heterozygous variants in ALFY might lead to microcephaly, a C-terminal fragment consisting of 1226 amino acids of both wild-type and variant human ALFY was expressed in developing Drosophila brains (Kadir et al., 2016). This approach showed that, while pupal brains expressing wild type human ALFY appeared normal, animals expressing mutant ALFY failed to eclose (enter adulthood) and showed brain sizes 40-60% smaller than brains expressing wild-type ALFY. Additionally, expression of mutant ALFY in the eye resulted in a severe rough eye phenotype, while eyes expressing wild-type ALFY appeared normal. Based on further in vivo and in vitro analyses, the authors proposed a mechanism in which the expression of mutant ALFY causes sustained Wnt signaling in radial glial cells, which thereby promotes symmetric division of apical progenitor cells at the expense of asymmetric division and the generation of basal progenitor cells. This disruption leads to fewer neurons and eventually microcephaly (Kadir et al., 2016). Taken together, these results indicate that heterozygous variants in ALFY lead to microcephaly, likely in a dominant-negative fashion, and provide a diagnosis for MCPH as well as revealing the mechanism by which ALFY mutations contribute to disease pathogenesis.

Drosophila have also been valuable for studying mechanisms of microcephaly without having to use ‘humanization’. For example, variants in WDR81 – a gene that encodes WD repeat-containing protein 81 – were recently identified in five families with associated progressive microcephaly (Cavallin et al., 2017). Individuals often initially presented with mild microcephaly but then progressed to more severe phenotypes as they aged (OFC z=−5 to −10 s.d.). Neurological development was severely disrupted in these individuals, and MRIs showed significant defects. Studies assessing the function of WDR81 in mutant human fibroblasts showed mitotic progression delay, and analogous studies in Drosophila neuronal stem cells (termed neuroblasts) with knockdown of Drosophila WDR81 revealed a similar cell cycle phenotype (Cavallin et al., 2017). Together, these results indicate that WDR81 is an essential component of the cell cycle machinery and support the diagnosis of microcephaly induced by variants of WDR81.

Variants in KATNB1, which encodes a regulatory subunit of the microtubule-severing enzyme katanin, were found in five individuals with complex brain malformations, including lissencephaly (defects in the development of brain folds) and microcephaly (Mishra-Gorur et al., 2014). Mutations in KATNB1 were shown to affect the mitotic spindle in dividing patient-derived fibroblasts, and corresponding studies in Drosophila showed reduced brain size after knockdown of the Drosophila ortholog of KATNB1, kat80. In addition, developing larval brains, which harbor neuroblasts that continuously divide to produce neurons for the entire adult brain, showed reduced numbers of neuroblasts with a significant delay in cell cycle progression and centrosome defects, correlating with the brain size reduction observed in third instar larvae. As mutations in both human and Drosophila KATNB1 resulted in reduced brain size, it was concluded that KATNB1 is likely associated with microcephaly in humans. Finally, it was noted that animals lacking kat80 demonstrated neuronal aborization defects in Drosophila, indicating that variants in KATNB1 could also affect the peripheral nervous system. Moving forward, it will be interesting to assess rescue by human WDR81 or KATNB1 and variants associated with human microcephaly in Drosophila models of asymmetric division and brain development.

Experiments using Drosophila have also helped to solve a case of a single individual affected by severe microcephaly; follow-up studies then led to the unraveling of the pathogenic mechanisms and to the discovery of many new genes associated with microcephaly in humans (Link et al., 2019; Yamamoto et al., 2014). A mutation in Ankle2 was recovered from a large mutagenesis screen in Drosophila designed to identify novel mutations associated with neurodevelopmental or neurodegenerative phenotypes (Yamamoto et al., 2014). To link Ankle2 to human disease, the human homolog (ANKLE2) was used to search the Baylor-Hopkins Center for Mendelian Genomics (BHCMG) exome database (Bamshad et al., 2012; Posey et al., 2019). A single individual was identified presenting with severe microcephaly (OFC z=−9 s.d.) and harboring compound heterozygous variants in ANKLE2. Both parents were carriers of the variants, fitting an autosomal recessive inheritance pattern. Mutations in Drosophila Ankle2 also gave rise to a small brain phenotype in larvae due to a reduction in cell division, fewer neuroblasts and an increase in cell death (Yamamoto et al., 2014). To verify that ANKLE2 was associated with human microcephaly, human wild-type and variant ANKLE2 cDNAs were expressed in Drosophila mutant for Ankle2. Wild-type human ANKLE2 rescued both lethality and brain size but the variants associated with human microcephaly did not, indicating that ANKLE2 mutations in both Drosophila and humans cause reduced brain size (Link et al., 2019). These results confirmed that ANKLE2 function is conserved, and that variants in ANKLE2 lead to primary microcephaly (MCPH16).

Additional studies showed that Ankle2 interacts with a kinase called Ballchen (VRK1 in humans) to regulate the asymmetric division of Drosophila neuroblasts (Link et al., 2019). It was also noted that the localization of key polarity proteins, including the PAR complex proteins aPKC, Par3 and Par6, was disrupted in dividing Ankle2 mutant neuroblasts. PAR proteins have long been established as essential proteins for regulating polarity in many cell types, including neuroblasts, and are known to affect cell fate decisions when disrupted (Betschinger et al., 2003; Rolls et al., 2003; Suzuki and Ohno, 2006). Defects in PAR complex localization in Ankle2 mutant animals resulted in defective asymmetric division and neuronal development, which contributed to their reduced brain volumes (Fig. 1). Using the human orthologs corresponding to the PAR complex and members of the ANKLE2 pathway, the BHCMG and Baylor Genetics databases were searched, and additional individuals with microcephaly and neuronal phenotypes were identified (Table 1). Although VRK1 has previously been associated with pontocerebellar hypoplasia (Gonzaga-Jauregui et al., 2013), PAR complex members had not previously been associated with microcephaly phenotypes. This approach revealed that the ability to pinpoint where Ankle2 and Ballchen/VRK1 fit into the neuroblast division pathway using Drosophila allowed the identification of essential genes associated with cell polarity and human neurological disease (Link et al., 2019). In the future, studies using Drosophila and mice to investigate how human variants in PAR complex proteins lead to neuronal phenotypes could provide crucial insight into disease pathogenesis.

Fig. 1.

Using Drosophila to identify an essential pathway that is required for brain development and is associated with human disease. (A,B) Drosophila third instar larval brains from (A) wild type and (B) severe Ankle2 mutants with associated microcephaly. Scale bars: 100 µm. (C) Ankle2 regulates asymmetric division by interacting with a kinase, Ballchen. Together, these proteins affect L(2)gl and aPKC activity, causing defects during division, including perturbing the localization of key asymmetric proteins that regulate cell fate. These include PAR complex proteins (aPKC, Par3 and Par6), which are localized apically (green) and control the localization of cell fate determinants on the basal side of the cell (purple). (D) Normal asymmetric division of a neuroblast (when PAR proteins and cell fate determinants are segregated properly) produces a stem cell that can self-renew and a ganglion mother cell that will eventually differentiate. (E) In the case of disrupted asymmetric division (when PAR proteins are not properly localized to the apical membrane), cell fate determinant localization is expanded and two ganglion mother cells, which go on to differentiate, are produced at the expense of a neuroblast. Alternatively, cell death can occur (not shown).

Fig. 1.

Using Drosophila to identify an essential pathway that is required for brain development and is associated with human disease. (A,B) Drosophila third instar larval brains from (A) wild type and (B) severe Ankle2 mutants with associated microcephaly. Scale bars: 100 µm. (C) Ankle2 regulates asymmetric division by interacting with a kinase, Ballchen. Together, these proteins affect L(2)gl and aPKC activity, causing defects during division, including perturbing the localization of key asymmetric proteins that regulate cell fate. These include PAR complex proteins (aPKC, Par3 and Par6), which are localized apically (green) and control the localization of cell fate determinants on the basal side of the cell (purple). (D) Normal asymmetric division of a neuroblast (when PAR proteins and cell fate determinants are segregated properly) produces a stem cell that can self-renew and a ganglion mother cell that will eventually differentiate. (E) In the case of disrupted asymmetric division (when PAR proteins are not properly localized to the apical membrane), cell fate determinant localization is expanded and two ganglion mother cells, which go on to differentiate, are produced at the expense of a neuroblast. Alternatively, cell death can occur (not shown).

Table 1.

Human orthologs corresponding to the ANKLE2 pathway and their associated phenotypes in humans

Human orthologs corresponding to the ANKLE2 pathway and their associated phenotypes in humans
Human orthologs corresponding to the ANKLE2 pathway and their associated phenotypes in humans

In related studies, ANKLE2 has been shown to interact with a Zika virus protein, NS4A (Shah et al., 2018). Expression of NS4A in Drosophila causes microcephaly phenotypes, while co-expression of human ANKLE2 rescues NS4A-induced phenotypes. These investigations showed that NS4A expression causes microcephaly, and that this protein inhibits the function of ANKLE2, providing a compelling mechanism for Zika virus-induced microcephaly. Furthermore, NS4A-induced phenotypes are more severe in animals heterozygous for Ankle2 mutations, suggesting that human variants in microcephaly loci such as ANKLE2 may enhance Zika virus-associated microcephaly.

In summary, studies using Drosophila to assess ANKLE2 function provided a diagnosis for the first patient, offered mechanisms of disease pathogenesis and allowed identification of additional patients with variants in the ANKLE2 pathway. Furthermore, these experiments demonstrate that both genetic and viral-induced microcephaly result from inhibition of the same evolutionarily conserved pathway and provide evidence that genetic variation in humans may affect phenotypes associated with environmental pathogens. Obviously, not all processes that lead to microcephaly in human will be conserved. Indeed, some genes that have been shown to cause primary microcephaly in human do not have an obvious Drosophila homolog, and it is not yet clear whether a functional equivalent exists in Drosophila.

When combined with human genetics, studies in Drosophila offer an efficient and effective approach that can aid both the diagnosis of disease and the elucidation of disease mechanisms. Although we have placed an emphasis on microcephaly here, Drosophila can be used for the study of many human developmental disorders. Mechanistic insights using Drosophila enable diagnoses for patients and often provide clues for therapeutic targets. As technologies advance with human genetics, model systems will be essential for assessing the function of the numerous variants associated with disease. Drosophila provides an opportunity to help diagnose cases with few affected individuals, as shown for ANKLE2, or in the case of de novo and dominant-negative mutations, such as those associated with ALFY/WDFY3. Furthermore, with established lines from the Drosophila Genetic Reference Panel (MacKay et al., 2012), the role of genetic variation and how it might affect phenotypes associated with rare human disease variants can be explored (Chow et al., 2016; Lavoy et al., 2018). These studies also highlight the fact that collaborations between patient families, clinicians, human geneticists and model system geneticists can and will continue to solve rare disease-related questions. In summary, a cooperative, multidisciplinary approach promotes a positive outcome for patients and advances our biological knowledge. Given that many thousands of diseases likely remain to be discovered, we anticipate that these discoveries are just the beginning of an exciting biological and therapeutic adventure.

We thank Angad Jolly for critical reading of the manuscript.

Funding

This work was supported by the National Institutes of Health/National Institute of Neurological Disorders and Stroke (F32NS092270 to N.L.) and jointly funded by a National Human Genome Research Institute and National Heart, Lung, and Blood Institute grant to the Baylor-Hopkins Center for Mendelian Genomics (UM1 HG006542 to J.R.L, and NIH R01GM067858 and NIH R24OD022005 to H.J.B.). N.L. is supported by the Howard Hughes Medical Institute and H.J.B. is an Investigator of the Howard Hughes Medical Institute. Deposited in PMC for release after 12 months.

Adams
,
D. R.
and
Eng
,
C. M.
(
2018
).
Next-generation sequencing to diagnose suspected genetic disorders
.
N. Engl. J. Med.
17
,
405
-
424
.
Amberger
,
J. S.
,
Bocchini
,
C. A.
,
Schiettecatte
,
F.
,
Scott
,
A. F.
and
Hamosh
,
A.
(
2015
).
OMIM.org: Online Mendelian Inheritance in Man (OMIM®), an Online catalog of human genes and genetic disorders
.
Nucleic Acids Res.
43
,
D789
-
D798
.
Ansar
,
M.
,
Chung
,
H.
,
Waryah
,
Y. M.
,
Makrythanasis
,
P.
,
Falconnet
,
E.
,
Rao
,
A. R.
,
Guipponi
,
M.
,
Narsani
,
A. K.
,
Fingerhut
,
R.
,
Santoni
,
F. A.
, et al. 
(
2018a
).
Visual impairment and progressive phthisis bulbi caused by recessive pathogenic variant in MARK3
.
Hum. Mol. Genet.
27
,
2703
-
2711
.
Ansar
,
M.
,
Chung
,
H.
,
Taylor
,
R. L.
,
Nazir
,
A.
,
Imtiaz
,
S.
,
Sarwar
,
M. T.
,
Manousopoulou
,
A.
,
Makrythanasis
,
P.
,
Saeed
,
S.
,
Falconnet
,
E.
, et al. 
(
2018b
).
Bi-allelic Loss-of-Function Variants in DNMBP Cause Infantile Cataracts
.
Am. J. Hum. Genet.
103
,
568
-
578
.
Bamshad
,
M. J.
,
Shendure
,
J. A.
,
Valle
,
D.
,
Hamosh
,
A.
,
Lupski
,
J. R.
,
Gibbs
,
R. A.
,
Boerwinkle
,
E.
,
Lifton
,
R. P.
,
Gerstein
,
M.
,
Gunel
,
M.
, et al. 
(
2012
).
The Centers for Mendelian Genomics: a new large-scale initiative to identify the genes underlying rare Mendelian conditions
.
Am. J. Med. Genet. A
158
,
1523
-
1525
.
Bamshad
,
M. J.
,
Nickerson
,
D. A.
and
Chong
,
J. X.
(
2019
).
Mendelian Gene Discovery: Fast and Furious with No End in Sight
.
Am. J. Hum. Genet.
105
,
448
-
455
.
Beaulieu
,
C. L.
,
Majewski
,
J.
,
Schwartzentruber
,
J.
,
Samuels
,
M. E.
,
Fernandez
,
B. A.
,
Bernier
,
F. P.
,
Brudno
,
M.
,
Knoppers
,
B.
,
Marcadier
,
J.
,
Dyment
,
D.
, et al. 
(
2014
).
FORGE Canada consortium: Outcomes of a 2-year national rare-disease gene-discovery project
.
Am. J. Hum. Genet.
94
,
809
-
817
.
Bellen
,
H. J.
and
Yamamoto
,
S.
(
2015
).
Morgan's legacy: fruit flies and the functional annotation of conserved genes
.
Cell
163
,
12
-
14
.
Bellen
,
H. J.
,
Wangler
,
M. F.
and
Yamamoto
,
S.
(
2019
).
The fruit fly at the interface of diagnosis and pathogenic mechanisms of rare and common human diseases
.
Hum. Mol. Genet.
28
,
R207
-
R214
.
Bellosta
,
P.
and
Soldano
,
A.
(
2019
).
Dissecting the genetics of autism spectrum disorders: A Drosophila perspective
.
Front. Physiol.
10
,
987
.
Betschinger
,
J.
,
Mechtler
,
K.
and
Knoblich
,
J. A.
(
2003
).
The Par complex directs asymmetric cell division by phosphorylating the cytoskeletal protein Lgl
.
Nature
422
,
326
-
330
.
Boycott
,
K. M.
,
Campeau
,
P. M.
,
Howley
,
H. E.
,
Pavlidis
,
P.
,
Rogic
,
S.
,
Oriel
,
C.
,
Berman
,
J. N.
,
Hamilton
,
R. M.
,
Hicks
,
G. G.
,
Lipshitz
,
H. D.
, et al. 
(
2020
).
The Canadian Rare Diseases Models and Mechanisms (RDMM) Network: Connecting Understudied Genes to Model Organisms
.
Am. J. Hum. Genet.
106
,
143
-
152
.
Brand
,
A. H.
and
Perrimon
,
N.
(
1993
).
Targeted gene expression as a means of altering cell fates and generating dominant phenotypes
.
Development
118
,
401
-
415
.
Buske
,
O. J.
,
Schiettecatte
,
F.
,
Hutton
,
B.
,
Dumitriu
,
S.
,
Misyura
,
A.
,
Huang
,
L.
,
Hartley
,
T.
,
Girdea
,
M.
,
Sobreira
,
N.
,
Mungall
,
C.
, et al. 
(
2015
).
The Matchmaker Exchange API: Automating Patient Matching Through the Exchange of Structured Phenotypic and Genotypic Profiles
.
Hum. Mutat.
36
,
922
-
927
.
Cavallin
,
M.
,
Rujano
,
M. A.
,
Bednarek
,
N.
,
Medina-Cano
,
D.
,
Bernabe Gelot
,
A.
,
Drunat
,
S.
,
Maillard
,
C.
,
Garfa-Traore
,
M.
,
Bole
,
C.
,
Nitschké
,
P.
, et al. 
(
2017
).
WDR81 mutations cause extreme microcephaly and impair mitotic progression in human fibroblasts and Drosophila neural stem cells
.
Brain
140
,
2597
-
2609
.
Caygill
,
E. E.
and
Brand
,
A. H.
(
2016
).
The GAL4 system: A versatile system for the manipulation and analysis of gene expression
.
Methods Mol. Biol.
1478
,
33
-
52
.
Chao
,
Y.-H.
,
Robak
,
L. A.
,
Xia
,
F.
,
Koenig
,
M. K.
,
Adesina
,
A.
,
Bacino
,
C. A.
,
Scaglia
,
F.
,
Bellen
,
H. J.
and
Wangler
,
M. F.
(
2016
).
Missense variants in the middle domain of DNM1L in cases of infantile encephalopathy alter peroxisomes and mitochondria when assayed in Drosophila
.
Hum. Mol. Genet.
25
,
1846
-
1856
.
Chao
,
H.-T.
,
Davids
,
M.
,
Burke
,
E.
,
Pappas
,
J. G.
,
Rosenfeld
,
J. A.
,
McCarty
,
A. J.
,
Davis
,
T.
,
Wolfe
,
L.
,
Toro
,
C.
,
Tifft
,
C.
, et al. 
(
2017
).
A Syndromic Neurodevelopmental Disorder Caused by De Novo Variants in EBF3
.
Am. J. Hum. Genet.
100
,
128
-
137
.
Chow
,
C. Y.
and
Reiter
,
L. T.
(
2017
).
Etiology of Human Genetic Disease on the Fly
.
Trends Genet.
33
,
391
-
398
.
Chow
,
C. Y.
,
Kelsey
,
K. J. P.
,
Wolfner
,
M. F.
and
Clark
,
A. G.
(
2016
).
Candidate genetic modifiers of retinitis pigmentosa identified by exploiting natural variation in Drosophila
.
Hum. Mol. Genet.
25
,
651
-
659
.
Chung
,
H.
,
Wangler
,
M. F.
,
Marcogliese
,
P. C.
,
Jo
,
J.
,
Ravenscroft
,
T. A.
,
Zuo
,
Z.
,
Duraine
,
L.
,
Sadeghzadeh
,
S.
,
Li-Kroeger
,
D.
,
Schmidt
,
R. E.
, et al. 
(
2020
).
Loss- or Gain-of-Function Mutations in ACOX1 Cause Axonal Loss via Different Mechanisms
.
Neuron
106
,
589
-
606.e6
.
Davis
,
F. P.
,
Nern
,
A.
,
Picard
,
S.
,
Reiser
,
M. B.
,
Rubin
,
G. M.
,
Eddy
,
S. R.
and
Henry
,
G. L.
(
2020
).
A genetic, genomic, and computational resource for exploring neural circuit function
.
eLife
9
,
e50901
.
Devakumar
,
D.
,
Bamford
,
A.
,
Ferreira
,
M. U.
,
Broad
,
J.
,
Rosch
,
R. E.
,
Groce
,
N.
,
Breuer
,
J.
,
Cardoso
,
M. A.
,
Copp
,
A. J.
,
Alexandre
,
P.
, et al. 
(
2018
).
Infectious causes of microcephaly: epidemiology, pathogenesis, diagnosis, and management
.
Lancet Infect. Dis.
18
,
PE1
-
E13
.
Diao
,
F.
,
Ironfield
,
H.
,
Luan
,
H.
,
Shropshire
,
W. C.
,
Ewer
,
J.
,
Marr
,
E.
,
Potter
,
C. J.
,
Landgraf
,
M.
and
White
,
B. H.
(
2015
).
Plug-and-play genetic access to drosophila cell types using exchangeable exon cassettes
.
Cell Rep.
10
,
1410
-
1421
.
Duffy
,
J. B.
(
2002
).
GAL4 system in Drosophila: A fly geneticist's Swiss army knife
.
Genesis
34
,
1
-
15
.
Filimonenko
,
M.
,
Isakson
,
P.
,
Finley
,
K. D.
,
Anderson
,
M.
,
Jeong
,
H.
,
Melia
,
T. J.
,
Bartlett
,
B. J.
,
Myers
,
K. M.
,
Birkeland
,
H. C. G.
,
Lamark
,
T.
, et al. 
(
2010
).
The Selective Macroautophagic Degradation of Aggregated Proteins Requires the PI3P-Binding Protein Alfy
.
Mol. Cell
38
,
265
-
279
.
Firth
,
H. V.
and
Wright
,
C. F.
(
2011
).
The Deciphering Developmental Disorders (DDD) study
.
Dev. Med. Child Neurol.
53
,
702
-
703
.
Frésard
,
L.
,
Smail
,
C.
,
Ferraro
,
N. M.
,
Teran
,
N. A.
,
Li
,
X.
,
Smith
,
K. S.
,
Bonner
,
D.
,
Kernohan
,
K. D.
,
Marwaha
,
S.
,
Zappala
,
Z.
, et al. 
(
2019
).
Identification of rare-disease genes using blood transcriptome sequencing and large control cohorts
.
Nat. Med.
25
,
911
-
919
.
Gallaud
,
E.
,
Pham
,
T.
and
Cabernard
,
C.
(
2017
).
Drosophila melanogaster Neuroblasts: A Model for Asymmetric Stem Cell Divisions
.
Results Probl. Cell Differ.
61
,
183
-
210
.
Gambarotto
,
D.
,
Pennetier
,
C.
,
Ryniawec
,
J. M.
,
Buster
,
D. W.
,
Gogendeau
,
D.
,
Goupil
,
A.
,
Nano
,
M.
,
Simon
,
A.
,
Blanc
,
D.
,
Racine
,
V.
, et al. 
(
2019
).
Plk4 Regulates Centriole Asymmetry and Spindle Orientation in Neural Stem Cells
.
Dev. Cell
50
,
11
-
24.e10
.
Gasser
,
S. M.
,
Lupski
,
J. R.
,
Le Cam
,
Y.
and
Menzel
,
O.
(
2012
).
Leap year: Rare day to highlight rare diseases
.
Nature
481
,
265
.
Gonzaga-Jauregui
,
C.
,
Lotze
,
T.
,
Jamal
,
L.
,
Penney
,
S.
,
Campbell
,
I. M.
,
Pehlivan
,
D.
,
Hunter
,
J. V.
,
Woodbury
,
S. L.
,
Raymond
,
G.
,
Adesina
,
A. M.
, et al. 
(
2013
).
Mutations in VRK1 associated with complex motor and sensory axonal neuropathy plus microcephaly
.
JAMA Neurol.
70
,
1491
-
1498
.
Halperin
,
D.
,
Kadir
,
R.
,
Perez
,
Y.
,
Drabkin
,
M.
,
Yogev
,
Y.
,
Wormser
,
O.
,
Berman
,
E. M.
,
Eremenko
,
E.
,
Rotblat
,
B.
,
Shorer
,
Z.
, et al. 
(
2019
).
SEC31A mutation affects ER homeostasis, causing a neurological syndrome
.
J. Med. Genet.
56
,
139
-
114
.
Harel
,
T.
,
Yoon
,
W. H.
,
Garone
,
C.
,
Gu
,
S.
,
Coban-Akdemir
,
Z.
,
Eldomery
,
M. K.
,
Posey
,
J. E.
,
Jhangiani
,
S. N.
,
Rosenfeld
,
J. A.
,
Cho
,
M. T.
, et al. 
(
2016
).
Recurrent De Novo and Biallelic Variation of ATAD3A, Encoding a Mitochondrial Membrane Protein, Results in Distinct Neurological Syndromes
.
Am. J. Hum. Genet.
99
,
831
-
845
.
Hirth
,
F.
and
Reichert
,
H.
(
1999
).
Conserved genetic programs in insect and mammalian brain development
.
BioEssays
21
,
677
-
684
.
Homem
,
C. C.
and
Knoblich
,
J. A.
(
2012
).
Drosophila neuroblasts: a model for stem cell biology
.
Development
139
,
4297
-
4310
.
Hubert
,
L.
,
Cannata Serio
,
M.
,
Villoing-Gaudé
,
L.
,
Boddaert
,
N.
,
Kaminska
,
A.
,
Rio
,
M.
,
Lyonnet
,
S.
,
Munnich
,
A.
,
Poirier
,
K.
,
Simons
,
M.
, et al. 
(
2020
).
De novo SCAMP5 mutation causes a neurodevelopmental disorder with autistic features and seizures
.
J. Med. Genet.
57
,
138
-
144
.
Jayaraman
,
D.
,
Bae
,
B. I.
and
Walsh
,
C. A.
(
2018
).
The Genetics of Primary Microcephaly
.
Annu. Rev. Genomics Hum. Genet.
19
,
177
-
200
.
Kadir
,
R.
,
Harel
,
T.
,
Markus
,
B.
,
Perez
,
Y.
,
Bakhrat
,
A.
,
Cohen
,
I.
,
Volodarsky
,
M.
,
Feintsein-Linial
,
M.
,
Chervinski
,
E.
,
Zlotogora
,
J.
, et al. 
(
2016
).
ALFY-Controlled DVL3 Autophagy Regulates Wnt Signaling, Determining Human Brain Size
.
PLoS Genet.
12
,
e1005919
.
Kanca
,
O.
,
Bellen
,
H. J.
and
Schnorrer
,
F.
(
2017
).
Gene tagging strategies to assess protein expression, localization, and function in Drosophila
.
Genetics
207
,
389
-
412
.
Kummeling
,
J.
,
Stremmelaar
,
D. E.
,
Raun
,
N.
,
Reijnders
,
M. R. F.
,
Willemsen
,
M. H.
,
Ruiterkamp-Versteeg
,
M.
,
Schepens
,
M.
,
Man
,
C. C. O.
,
Gilissen
,
C.
,
Cho
,
M. T.
, et al. 
(
2020
).
Characterization of SETD1A haploinsufficiency in humans and Drosophila defines a novel neurodevelopmental syndrome
.
Mol. Psychiatry
Lavoy
,
S.
,
Chittoor-Vinod
,
V. G.
,
Chow
,
C. Y.
and
Martin
,
I.
(
2018
).
Genetic modifiers of neurodegeneration in a drosophila model of parkinson's disease
.
Genetics
209
,
1345
-
1356
.
Lin
,
G.
,
Lee
,
P.-T.
,
Chen
,
K.
,
Mao
,
D.
,
Tan
,
K. L.
,
Zuo
,
Z.
,
Lin
,
W.-W.
,
Wang
,
L.
and
Bellen
,
H. J.
(
2018
).
Phospholipase PLA2G6, a Parkinsonism-Associated Gene, Affects Vps26 and Vps35, Retromer Function, and Ceramide Levels, Similar to α-Synuclein Gain
.
Cell Metab.
28
,
605
-
618.e6
.
Link
,
N.
,
Chung
,
H.
,
Jolly
,
A.
,
Withers
,
M.
,
Tepe
,
B.
,
Arenkiel
,
B. R.
,
Shah
,
P. S.
,
Krogan
,
N. J.
,
Aydin
,
H.
,
Geckinli
,
B. B.
, et al. 
(
2019
).
Mutations in ANKLE2, a ZIKA Virus Target, Disrupt an Asymmetric Cell Division Pathway in Drosophila Neuroblasts to Cause Microcephaly
.
Dev. Cell
51
,
713
-
729.e6
.
Liu
,
L.
,
MacKenzie
,
K. R.
,
Putluri
,
N.
,
Maletić-Savatić
,
M.
and
Bellen
,
H. J.
(
2017
).
The Glia-Neuron Lactate Shuttle and Elevated ROS Promote Lipid Synthesis in Neurons and Lipid Droplet Accumulation in Glia via APOE/D
.
Cell Metab.
26
,
719
-
737
.
Liu
,
P.
,
Meng
,
L.
,
Normand
,
E. A.
,
Xia
,
F.
,
Song
,
X.
,
Ghazi
,
A.
,
Rosenfeld
,
J.
,
Magoulas
,
P. L.
,
Braxton
,
A.
,
Ward
,
P.
, et al. 
(
2019
).
Reanalysis of clinical exome sequencing data
.
N. Engl. J. Med.
380
,
2478
-
2480
.
Lucas
,
E. P.
and
Raff
,
J. W.
(
2007
).
Maintaining the proper connection between the centrioles and the pericentriolar matrix requires Drosophila centrosomin
.
J. Cell Biol.
178
,
725
-
732
.
Luo
,
X.
,
Rosenfeld
,
J. A.
,
Yamamoto
,
S.
,
Harel
,
T.
,
Zuo
,
Z.
,
Hall
,
M.
,
Wierenga
,
K. J.
,
Pastore
,
M. T.
,
Bartholomew
,
D.
,
Delgado
,
M. R.
, et al. 
(
2017
).
Clinically severe CACNA1A alleles affect synaptic function and neurodegeneration differentially
.
PLoS Genet.
13
,
e1006905
.
MacKay
,
T. F. C.
,
Richards
,
S.
,
Stone
,
E. A.
,
Barbadilla
,
A.
,
Ayroles
,
J. F.
,
Zhu
,
D.
,
Casillas
,
S.
,
Han
,
Y.
,
Magwire
,
M. M.
,
Cridland
,
J. M.
, et al. 
(
2012
).
The Drosophila melanogaster Genetic Reference Panel
.
Nature
482
,
173
-
178
.
Marcogliese
,
P. C.
,
Shashi
,
V.
,
Spillmann
,
R. C.
,
Stong
,
N.
,
Rosenfeld
,
J. A.
,
Koenig
,
M. K.
,
Martínez-Agosto
,
J. A.
,
Herzog
,
M.
,
Chen
,
A. H.
,
Dickson
,
P. I.
, et al. 
(
2018
).
IRF2BPL Is Associated with Neurological Phenotypes
.
Am. J. Hum. Genet.
103
,
245
-
260
.
Mishra-Gorur
,
K.
,
Çağlayan
,
A. O.
,
Schaffer
,
A. E.
,
Chabu
,
C.
,
Henegariu
,
O.
,
Vonhoff
,
F.
,
Akgümüş
,
G. T.
,
Nishimura
,
S.
,
Han
,
W.
,
Tu
,
S.
, et al. 
(
2014
).
Mutations in KATNB1 cause complex cerebral malformations by disrupting asymmetrically dividing neural progenitors
.
Neuron
84
,
1226
-
1239
.
Muir
,
A. M.
,
Cohen
,
J. L.
,
Sheppard
,
S. E.
,
Guttipatti
,
P.
,
Lo
,
T. Y.
,
Weed
,
N.
,
Doherty
,
D.
,
DeMarzo
,
D.
,
Fagerberg
,
C. R.
,
Kjærsgaard
,
L.
, et al. 
(
2020
).
Bi-allelic Loss-of-Function Variants in NUP188 Cause a Recognizable Syndrome Characterized by Neurologic, Ocular, and Cardiac Abnormalities
.
Am. J. Hum. Genet.
106
,
623
-
631
.
Naveed
,
M.
,
Kazmi
,
S. K.
,
Amin
,
M.
,
Asif
,
Z.
,
Islam
,
U.
,
Shahid
,
K.
and
Tehreem
,
S.
(
2018
).
Comprehensive review on the molecular genetics of autosomal recessive primary microcephaly (MCPH)
.
Genet. Res.
100
,
e7
.
Ngo
,
K. J.
,
Rexach
,
J. E.
,
Lee
,
H.
,
Petty
,
L. E.
,
Perlman
,
S.
,
Valera
,
J. M.
,
Deignan
,
J. L.
,
Mao
,
Y.
,
Aker
,
M.
,
Posey
,
J. E.
, et al. 
(
2020
).
A diagnostic ceiling for exome sequencing in cerebellar ataxia and related neurological disorders
.
Hum. Mutat.
41
,
487
-
501
.
Nixon
,
K. C. J.
,
Rousseau
,
J.
,
Stone
,
M. H.
,
Sarikahya
,
M.
,
Ehresmann
,
S.
,
Mizuno
,
S.
,
Matsumoto
,
N.
,
Miyake
,
N.
,
Baralle
,
D.
,
McKee
,
S.
, et al. 
(
2019
).
A Syndromic Neurodevelopmental Disorder Caused by Mutations in SMARCD1, a Core SWI/SNF Subunit Needed for Context-Dependent Neuronal Gene Regulation in Flies
.
Am. J. Hum. Genet.
104
,
596
-
610
.
Oláhová
,
M.
,
Yoon
,
W. H.
,
Thompson
,
K.
,
Jangam
,
S.
,
Fernandez
,
L.
,
Davidson
,
J. M.
,
Kyle
,
J. E.
,
Grove
,
M. E.
,
Fisk
,
D. G.
,
Kohler
,
J. N.
, et al. 
(
2018
).
Biallelic Mutations in ATP5F1D, which Encodes a Subunit of ATP Synthase, Cause a Metabolic Disorder
.
Am. J. Hum. Genet.
102
,
494
-
504
.
Olsen
,
S. R.
and
Wilson
,
R. I.
(
2008
).
Cracking neural circuits in a tiny brain: new approaches for understanding the neural circuitry of Drosophila
.
Trends Neurosci.
31
,
512
-
520
.
Owings
,
K. G.
,
Lowry
,
J. B.
,
Bi
,
Y.
,
Might
,
M.
and
Chow
,
C. Y.
(
2018
).
Transcriptome and functional analysis in a Drosophila model of NGLY1 deficiency provides insight into therapeutic approaches
.
Hum. Mol. Genet.
27
,
1055
-
1066
.
Pires-daSilva
,
A.
and
Sommer
,
R. J.
(
2003
).
The evolution of signalling pathways in animal development
.
Nat. Rev. Genet.
4
,
39
-
49
.
Pirozzi
,
F.
,
Nelson
,
B.
and
Mirzaa
,
G.
(
2018
).
From microcephaly to megalencephaly: Determinants of brain size
.
Dialogues Clin. Neurosci.
20
,
267
-
282
.
Posey
,
J. E.
,
O'Donnell-Luria
,
A. H.
,
Chong
,
J. X.
,
Harel
,
T.
,
Jhangiani
,
S. N.
,
Coban Akdemir
,
Z. H.
,
Buyske
,
S.
,
Pehlivan
,
D.
,
Carvalho
,
C. M. B.
,
Baxter
,
S.
, et al. 
(
2019
).
Insights into genetics, human biology and disease gleaned from family based genomic studies
.
Genet. Med.
21
,
798
-
812
.
Poulton
,
J. S.
,
Cuningham
,
J. C.
and
Peifer
,
M.
(
2017
).
Centrosome and spindle assembly checkpoint loss leads to neural apoptosis and reduced brain size
.
J. Cell Biol.
216
,
1255
-
1265
.
Ramdas Nair
,
A.
,
Singh
,
P.
,
Salvador Garcia
,
D.
,
Rodriguez-Crespo
,
D.
,
Egger
,
B.
and
Cabernard
,
C.
(
2016
).
The Microcephaly-Associated Protein Wdr62/CG7337 Is Required to Maintain Centrosome Asymmetry in Drosophila Neuroblasts
.
Cell Rep.
14
,
1100
-
1113
.
Ramoni
,
R. B.
,
Mulvihill
,
J. J.
,
Adams
,
D. R.
,
Allard
,
P.
,
Ashley
,
E. A.
,
Bernstein
,
J. A.
,
Gahl
,
W. A.
,
Hamid
,
R.
,
Loscalzo
,
J.
,
McCray
,
A. T.
, et al. 
(
2017
).
The Undiagnosed Diseases Network: Accelerating Discovery about Health and Disease
.
Am. J. Hum. Genet.
100
,
185
-
192
.
Reiter
,
L. T.
,
Potocki
,
L.
,
Chien
,
S.
,
Gribskov
,
M.
and
Bier
,
E.
(
2001
).
A systematic analysis of human disease-associated gene sequences in Drosophila melanogaster
.
Genome Res.
11
,
1114
-
1125
.
Rolls
,
M. M.
,
Albertson
,
R.
,
Shih
,
H.-P.
,
Lee
,
C.-Y.
and
Doe
,
C. Q.
(
2003
).
Drosophila aPKC regulates cell polarity and cell proliferation in neuroblasts and epithelia
.
J. Cell Biol.
163
,
1089
-
1098
.
Rujano
,
M. A.
,
Sanchez-Pulido
,
L.
,
Pennetier
,
C.
,
Le Dez
,
G.
and
Basto
,
R.
(
2013
).
The microcephaly protein Asp regulates neuroepithelium morphogenesis by controlling the spatial distribution of myosin II
.
Nat. Cell Biol.
15
,
1294
-
1306
.
Sáchez-Soriano
,
N.
,
Tea
,
G.
,
Whitington
,
P.
and
Prokop
,
A.
(
2007
).
Drosophila as a genetic and cellular model for studies on axonal growth
.
Neural Dev.
2
,
9
.
Sawyer
,
S. L.
,
Hartley
,
T.
,
Dyment
,
D. A.
,
Beaulieu
,
C. L.
,
Schwartzentruber
,
J.
,
Smith
,
A.
,
Bedford
,
H. M.
,
Bernard
,
G.
,
Bernier
,
F. P.
,
Brais
,
B.
, et al. 
(
2016
).
Utility of whole-exome sequencing for those near the end of the diagnostic odyssey: Time to address gaps in care
.
Clin. Genet.
89
,
275
-
284
.
Schoborg
,
T.
,
Zajac
,
A. L.
,
Fagerstrom
,
C. J.
,
Guillen
,
R. X.
and
Rusan
,
N. M.
(
2015
).
An Asp-CaM complex is required for centrosome-pole cohesion and centrosome inheritance in neural stem cells
.
J. Cell Biol.
211
,
987
-
998
.
Schoborg
,
T. A.
,
Smith
,
S. L.
,
Smith
,
L. N.
,
Douglas Morris
,
H.
and
Rusan
,
N. M.
(
2019
).
Micro-computed tomography as a platform for exploring Drosophila development
.
Development
146
,
dev176685
.
Şentürk
,
M.
,
Lin
,
G.
,
Zuo
,
Z.
,
Mao
,
D.
,
Watson
,
E.
,
Mikos
,
A. G.
and
Bellen
,
H. J.
(
2019
).
Ubiquilins regulate autophagic flux through mTOR signalling and lysosomal acidification
.
Nat. Cell Biol.
21
,
384
-
396
.
Shah
,
P. S.
,
Link
,
N.
,
Jang
,
G. M.
,
Sharp
,
P. P.
,
Zhu
,
T.
,
Swaney
,
D. L.
,
Johnson
,
J. R.
,
Von Dollen
,
J.
,
Ramage
,
H. R.
,
Satkamp
,
L.
, et al. 
(
2018
).
Comparative Flavivirus-Host Protein Interaction Mapping Reveals Mechanisms of Dengue and Zika Virus Pathogenesis
.
Cell
175
,
1931
-
1945.e18
.
Simonsen
,
A.
,
Birkeland
,
H. C. G.
,
Gillooly
,
D. J.
,
Mizushima
,
N.
,
Kuma
,
A.
,
Yoshimori
,
T.
,
Slagsvold
,
T.
,
Brech
,
A.
and
Stenmark
,
H.
(
2004
).
Alfy, a novel FYVE-domain-containing protein associated with protein granules and autophagic membranes
.
J. Cell Sci.
117
,
4239
-
4251
.
Singh
,
P.
,
Ramdas Nair
,
A.
and
Cabernard
,
C.
(
2014
).
The centriolar protein Bld10/Cep135 is required to establish centrosome asymmetry in drosophila neuroblasts
.
Curr. Biol.
24
,
1548
-
1555
.
Sobreira
,
N.
,
Schiettecatte
,
F.
,
Valle
,
D.
and
Hamosh
,
A.
(
2015
).
GeneMatcher: A Matching Tool for Connecting Investigators with an Interest in the Same Gene
.
Hum. Mutat.
36
,
928
-
930
.
Splinter
,
K.
,
Adams
,
D. R.
,
Bacino
,
C. A.
,
Bellen
,
H. J.
,
Bernstein
,
J. A.
,
Cheatle-Jarvela
,
A. M.
,
Eng
,
C. M.
,
Esteves
,
C.
,
Gahl
,
W. A.
,
Hamid
,
R.
, et al. 
(
2018
).
Effect of genetic diagnosis on patients with previously undiagnosed disease
.
N. Engl. J. Med.
379
,
2131
-
2139
.
Straub
,
J.
,
Konrad
,
E. D. H.
,
Grüner
,
J.
,
Toutain
,
A.
,
Bok
,
L. A.
,
Cho
,
M. T.
,
Crawford
,
H. P.
,
Dubbs
,
H.
,
Douglas
,
G.
,
Jobling
,
R.
, et al. 
(
2018
).
Missense Variants in RHOBTB2 Cause a Developmental and Epileptic Encephalopathy in Humans, and Altered Levels Cause Neurological Defects in Drosophila
.
Am. J. Hum. Genet.
102
,
44
-
57
.
Suzuki
,
A.
and
Ohno
,
S.
(
2006
).
The PAR-aPKC system: lessons in polarity
.
J. Cell Sci.
119
,
979
-
987
.
Taruscio
,
D.
,
Groft
,
S. C.
,
Cederroth
,
H.
,
Melegh
,
B.
,
Lasko
,
P.
,
Kosaki
,
K.
,
Baynam
,
G.
,
McCray
,
A.
and
Gahl
,
W. A.
(
2015
).
Undiagnosed Diseases Network International (UDNI): White paper for global actions to meet patient needs
.
Mol. Genet. Metab.
116
,
223
-
225
.
Wangler
,
M. F.
,
Yamamoto
,
S.
,
Chao
,
H.-T.
,
Posey
,
J. E.
,
Westerfield
,
M.
,
Postlethwait
,
J.
,
Hieter
,
P.
,
Boycott
,
K. M.
,
Campeau
,
P. M.
,
Bellen
,
H. J.
, et al. 
(
2017a
).
Model Organisms Facilitate Rare Disease Diagnosis and Therapeutic Research
.
Genetics
207
,
9
-
27
.
Wangler
,
M. F.
,
Chao
,
Y.-H.
,
Bayat
,
V.
,
Giagtzoglou
,
N.
,
Shinde
,
A. B.
,
Putluri
,
N.
,
Coarfa
,
C.
,
Donti
,
T.
,
Graham
,
B. H.
,
Faust
,
J. E.
, et al. 
(
2017b
).
Peroxisomal biogenesis is genetically and biochemically linked to carbohydrate metabolism in Drosophila and mouse
.
PLoS Genet.
13
,
e1006825
.
Wijsman
,
E. M.
(
2012
).
The role of large pedigrees in an era of high-Throughput sequencing
.
Hum. Genet.
131
,
1555
-
1563
.
Yamamoto
,
S.
,
Jaiswal
,
M.
,
Charng
,
W.-L.
,
Gambin
,
T.
,
Karaca
,
E.
,
Mirzaa
,
G.
,
Wiszniewski
,
W.
,
Sandoval
,
H.
,
Haelterman
,
N. A.
,
Xiong
,
B.
, et al. 
(
2014
).
A drosophila genetic resource of mutants to study mechanisms underlying human genetic diseases
.
Cell
159
,
200
-
214
.
Yoon
,
W. H.
,
Sandoval
,
H.
,
Nagarkar-Jaiswal
,
S.
,
Jaiswal
,
M.
,
Yamamoto
,
S.
,
Haelterman
,
N. A.
,
Putluri
,
N.
,
Putluri
,
V.
,
Sreekumar
,
A.
,
Tos
,
T.
, et al. 
(
2017
).
Loss of Nardilysin, a Mitochondrial Co-chaperone for α-Ketoglutarate Dehydrogenase, Promotes mTORC1 Activation and Neurodegeneration
.
Neuron
93
,
115
-
131
.
Zheng
,
Z.
,
Lauritzen
,
J. S.
,
Perlman
,
E.
,
Robinson
,
C. G.
,
Nichols
,
M.
,
Milkie
,
D.
,
Torrens
,
O.
,
Price
,
J.
,
Fisher
,
C. B.
,
Sharifi
,
N.
, et al. 
(
2018
).
A Complete Electron Microscopy Volume of the Brain of Adult Drosophila melanogaster
.
Cell
174
,
730
-
743.e22
.

Competing interests

The authors declare no competing or financial interests.