<<

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

1 Drosophila models of pathogenic copy-number variant show global and

2 non-neuronal defects during development

3

4 Tanzeen Yusuff1,4, Matthew Jensen1,4, Sneha Yennawar1,4, Lucilla Pizzo1, Siddharth

5 Karthikeyan1, Dagny J. Gould1, Avik Sarker1, Yurika Matsui1,2, Janani Iyer1, Zhi-Chun Lai1,2,

6 and Santhosh Girirajan1,3*

7

8 1. Department of Biochemistry and Molecular Biology, Pennsylvania State University,

9 University Park, PA 16802

10 2. Department of Biology, Pennsylvania State University, University Park, PA 16802

11 3. Department of Anthropology, Pennsylvania State University, University Park, PA 16802

12

13 4 contributed equally to work

14

15

16 *Correspondence:

17 Santhosh Girirajan, MBBS, PhD

18 205A Life Sciences Building

19 Pennsylvania State University

20 University Park, PA 16802

21 E-mail: [email protected]

22 Phone: 814-865-0674

23

1

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

24 ABSTRACT

25 While rare pathogenic CNVs are associated with both neuronal and non-neuronal ,

26 functional studies evaluating these regions have focused on the molecular basis of neuronal

27 defects. We report a systematic functional analysis of non-neuronal phenotypes for 59 homologs

28 of genes within ten CNVs and 20 neurodevelopmental genes in Drosophila. Using wing-specific

29 knockdown of 136 RNA interference lines, we identify phenotypes in 72/79 homologs including

30 six lines with lethality and 21 lines with severe phenotypes. We find no correlation between

31 severity of these phenotypes and neuronal defects due to eye-specific knockdown. We observe

32 disruptions in proliferation and apoptosis for 23/27 homologs, and altered Wnt, Hedgehog

33 and Notch signaling for 9/14 homologs, including AATF/Aatf, PPP4C/Pp4-19C, and

34 KIF11/Klp61F, validated with differences in tissue-specific expression and network

35 connectivity. Our findings suggest that multiple genes within each CNV differentially affect both

36 global and tissue-specific developmental processes, contributing to non-neuronal phenotypes of

37 CNV disorders.

38

39

2

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

40 INTRODUCTION

41 Rare copy-number variants (CNVs), or deletions and duplications in the genome, are associated

42 with neurodevelopmental disorders such as autism, intellectual disability (ID), and

43 schizophrenia1,2. While dosage alteration of CNV regions contribute predominantly to defects in

44 nervous system development, several CNV disorders also lead to early developmental features

45 involving other organ systems3,4, including cardiac defects5,6, kidney malformations7,

46 craniofacial features3, and skeletal abnormalities8. In fact, an overall survey of ten rare disease-

47 associated CNVs among individuals within the DECIPHER database9 showed a wide range of

48 non-neuronal phenotypes across multiple organ systems for each CNV disorder (Fig. 1). For

49 example, the 1q21.1 deletion causes variable expression of multiple neuronal and non-neuronal

50 phenotypes, including developmental delay, autism, and schizophrenia as well as craniofacial

51 features, cataracts, cardiac defects, and skeletal abnormalities10–12. Additionally, while the

52 7q11.23 deletion associated with Williams-Beuren syndrome (WBS) causes neuropsychiatric and

53 behavioral features, other non-neuronal phenotypes, including supravalvular aortic stenosis,

54 auditory defects, hypertension, diabetes mellitus, and musculoskeletal and connective tissue

55 anomalies, are also observed among the deletion carriers13. In fact, individual genes within the

56 WBS region are associated with specific features of the deletion, such as ELN and supravalvular

57 aortic stenosis14, STX1A and impaired glucose tolerance15, LIMK1 and impaired visuospatial

58 abilities16, and GTF2IRD1 and craniofacial abnormalities17. Furthermore, TBX1 was identified to

59 cause the pharyngeal arch cardiac defects associated with the 22q11.2 deletion (DiGeorge

60 syndrome)18, while HNF1B within the 17q12 deletion region was identified as the causative

61 for kidney defects associated with the deletion19,20. However, candidate genes for a majority of

62 non-neuronal phenotypes have not been identified for several rare CNV disorders, in particular

3

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

63 for CNVs associated with variably-expressive phenotypes such as the 1q21.1 deletion and the

64 16p11.2 deletion21,22. Additionally, affected individuals who carry disruptive in

65 neurodevelopmental genes from recent sequencing studies have also been documented to

66 manifest non-neuronal phenotypes. For example, individuals with loss-of-function mutations in

67 the autism-associated gene CHD8 present with gastrointestinal problems, tall stature, and

68 craniofacial features in addition to neuropsychiatric features23, while mutations in the

69 microcephaly-associated gene KIF11 also lead to congenital lymphedema and retinopathy24.

70 Despite the importance of identifying genes within CNVs that contribute towards non-

71 neuronal phenotypes, functional studies of CNV genes have primarily focused on detailed

72 assessments of neuronal phenotypes in model systems. For example, mouse models generated for

73 the 16p11.2 deletion exhibited post-natal lethality, reduced brain size and neural progenitor cell

74 count, motor and habituation defects, synaptic defects, and behavioral defects25–27. Similarly,

75 mouse models for the 3q29 deletion showed decreased weight and brain size, increased

76 locomotor activity and startle response, and decreased spatial learning and memory28,29.

77 However, fewer studies have focused on detailed evaluation of non-neuronal phenotypes in

78 functional models of CNV disorders. For example, Arbogast and colleagues evaluated obesity

79 and metabolic changes in 16p11.2 deletion mice, which showed reduced weight and impaired

80 adipogenesis30. While Haller and colleagues showed that mice with knockdown of MAZ, a gene

81 within the 16p11.2 deletion region, contribute to the genitourinary defects observed in

82 individuals with the deletion31, mouse studies on other homologs of 16p11.2 genes, including

83 TAOK2, KCTD13, and MAPK3, have only focused on assessing neuronal defects32–36.

84 Furthermore, Dickinson and colleagues reported a high-throughput analysis of essential genes in

85 mice and identified both neuronal and non-neuronal phenotypes for individual gene knockouts,

4

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

86 including more than 400 genes that lead to lethality37. While these efforts aided in implicating

87 novel genes with human disease, our understanding of how genes associated with

88 neurodevelopmental disorders contribute towards non-neuronal phenotypes is still limited.

89 Therefore, a large-scale analysis of non-neuronal phenotypes is necessary to identify specific

90 candidate genes within CNV regions and associated biological mechanisms that contribute

91 towards these phenotypes.

92 is an excellent model system to evaluate homologs of

93 neurodevelopmental genes, as many developmental processes and signaling pathways are

94 conserved between and flies38. In fact, over 75% of human disease genes have homologs

95 in Drosophila, including many genes involved in cellular signaling processes39,40. We recently

96 examined the contributions of individual Drosophila homologs of 28 genes within the 16p11.2

97 and 3q29 deletion regions towards specific neurodevelopmental phenotypes, including rough eye

98 phenotypes and defects in climbing ability, axon targeting, neuromuscular junction, and dendritic

99 arborization41,42. While these findings implicated multiple genes within each CNV region

100 towards neuronal phenotypes, the conserved role of these genes towards non-neuronal

101 phenotypes is not well understood. The Drosophila wing is an effective model system to

102 evaluate such developmental phenotypes, as key components of conserved signaling pathways,

103 such as Notch, epidermal growth factor (EGFR), Hegdehog, and Wnt pathways, were

104 identified using fly wing models43–49. For example, Wu and colleagues showed that

105 overexpression of the Drosophila homolog for UBE3A, associated with Angelman syndrome,

106 leads to abnormal wing and eye morphology defects50. Furthermore, Drosophila mutant screens

107 for developmental phenotypes, including wing defects, were used to identify conserved genes for

108 several human genetic diseases, including Charcot-Marie-Tooth disease and syndromic

5

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

109 microcephaly51. Kochinke and colleagues also recently performed a large-scale screening of ID-

110 associated genes, and found an enrichment of wing trichome density and missing vein

111 phenotypes in ID genes compared to control gene sets52. Hence, the fly wing provides a model

112 system that is ideal for evaluating the contributions of individual homologs of CNV genes

113 towards cellular and developmental phenotypes.

114 In this study, we tested the non-neuronal phenotypes of 79 fly homologs of human genes

115 within ten pathogenic CNV regions and genes associated with neurodevelopmental disorders.

116 We observed a wide range of robust qualitative and quantitative adult wing phenotypes among

117 the 136 RNA interference (RNAi) lines tested in our study, including size defects, ectopic and

118 missing veins, severe wrinkling, and lethality. Further analysis of cellular phenotypes revealed

119 disruptions in conserved developmental processes in the larval imaginal wing disc, including

120 altered levels of cell proliferation and apoptosis as well as altered expression patterns in the Wnt,

121 Hedgehog, and Notch signaling pathways. However, we found no correlation in the severity of

122 phenotypes observed with wing and eye-specific knockdown. Our findings were further

123 supported by differences in expression patterns and network connectivity of human CNV genes

124 across different tissues. Our analysis emphasizes the importance of multiple genes within each

125 CNV region towards both global and tissue-specific developmental processes, potentially

126 accounting for the non-neuronal phenotypes associated with pathogenic CNVs.

127

6

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

128 RESULTS

129 Wing-specific knockdown of fly homologs of CNV genes show non-neuronal phenotypes

130 Using an RNAi based analysis driven by the bxMS1096-GAL4 wing-specific driver, we tested a

131 total of 136 RNAi lines for 59 homologs of genes within pathogenic CNV regions (chromosomal

132 locations 1q21.1, 3q29, 7q11.23, 15q11.2, 15q13.3, 16p11.2, distal 16p11.2, 16p12.1, 16p13.11,

133 and 17q12) and 20 homologs of genes associated with neurodevelopmental disorders (Supp.

134 Data 1). Fly homologs of these genes were identified using the DIOPT orthology prediction

135 tool53. We list both the human gene name and the fly gene name for each tested gene as HUMAN

136 GENE/Fly gene (i.e. KCTD13/CG10465) as well as the human CNV region for context at first

137 instance. We scored 20-25 adult wings for five distinct wing phenotypes in each non-lethal

138 RNAi line, including wrinkled wing, discoloration, ectopic veins, missing veins, and bristle

139 planar polarity phenotypes (Fig. 2A; Supp. Data 2). We first categorized each wing

140 based on their severity and performed k-means clustering analysis to categorize each RNAi line

141 by their overall phenotype severity (Fig. 2B-C). We observed four clusters of RNAi lines: 75

142 lines with no observable qualitative phenotypes (55.2%), 24 lines with mild phenotypes (17.7%),

143 10 lines with moderate phenotypes (7.4%), 21 lines with severe phenotypes (15.4%), and 6 lines

144 with lethal phenotypes (4.4%), including ACACA/ACC within 17q12, DLG1/dlg1 within 3q29,

145 and STX1A/Syx1A within 7q11.23 (Fig. 2B-D; Supp. Data 2). We observed severe wrinkled

146 wing phenotypes for 13/79 fly homologs, including PPP4C/Pp4-19C within 16p11.2,

147 ATXN2L/Atx2 within distal 16p11.2, AATF/Aatf within 17q12, and MFI2/Tsf2 within 3q29 (Fig.

148 3A-B, Supp. Data 3). Interestingly, seven out of ten CNV regions contained at least one

149 homolog that showed lethality or severe wing phenotypes, and five CNV regions (3q29, 16p11.2,

150 distal 16p11.2, 16p12.1, and 17q12) had multiple homologs showing lethality or severe wing

7

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

151 phenotypes (Fig. 3A, Supp. Data 3). For example, RNAi lines for both UQCRC2/UQCR-C2 and

152 POLR3E/Sin within 16p12.1 showed lethality. Within the 3q29 region, NCBP2/Cbp20 and

153 MFI2/Tsf2 showed severe phenotypes while DLG1/dlg1 showed lethality. In contrast, 12/20

154 known neurodevelopmental genes showed no observable wing phenotypes, suggesting that these

155 genes could be responsible for neuronal-specific phenotypes (Fig. 3B, Supp. Data 3). We note

156 that 18/79 fly homologs showed discordant phenotypes between two or more RNAi lines for the

157 same gene, which could be due to differences in expression of the RNAi construct among these

158 lines (Supp. Data 3).

159 Certain qualitative phenotypes exhibited higher frequency in males compared to females.

160 For example, discoloration (87 lines in males compared to 56 lines in females; p=1.315×10-4,

161 two-tailed Fisher’s exact test) and missing vein phenotypes (92 lines in males compared to 29

162 lines in females; p=2.848×10-16, two-tailed Fisher’s exact test) at any degree of severity were

163 more commonly observed in males than females (Supp. Data 2). In particular, 25/92 lines in

164 males (compared to 1/29 in females) showed a total loss of the anterior crossvein (ACV) (Supp.

165 Data 2). We further identified 17 RNAi lines that were lethal in males with wing-specific

166 knockdown of fly homologs. While higher frequencies of wing phenotypes in males could be

167 due to a sex-specific bias of developmental phenotypes, the increased severity we observed in

168 males is most likely due to a stronger RNAi knockdown caused by an X-linked dosage

169 compensation, as the bxMS1096-GAL4 driver is inserted on the fly X chromosome54,55.

170 Next, we measured the total adult wing area and the lengths of six veins (longitudinal L2,

171 L3, L4, L5, ACV, and posterior crossvein or PCV) in the adult wing for each of the tested RNAi

172 lines that did not show lethality (or severe wrinkled phenotypes for vein length measurements)

173 (Fig. 4A). Overall, we identified significant wing measurement changes for 89 RNAi lines

8

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

174 compared to controls, which included lines that did not have an observable qualitative wing

175 phenotype (Fig. 2D). A summary of L3 vein lengths is presented in Fig. 4B, and the

176 measurements for the remaining five veins are presented in Supp. Figure 1 and Supp. Data. 2.

177 We found that 33/61 of the homologs (54%) showed significant changes in L3 vein length,

178 including 20 homologs with longer vein lengths and 13 homologs with shorter vein lengths

179 (Supp. Data 3). Additionally, 41/74 of the fly homologs (55%) showed changes in wing area

180 (Supp. Data 3), including 36 homologs which showed smaller wing areas and five homologs

181 showed larger wing areas compared to controls (Supp. Data 3). For example, both homologs of

182 genes within 1q21.1 region, BCL9/lgs and FMO5/Fmo-2, showed decreased wing area and vein

183 length, potentially mirroring the reduced body length phenotype observed in mouse models of

184 the deletion56 (Fig. 4B-C). In addition, PAK2/Pak within 3q29, TBX1/org-1 within 22q11.2,

185 autism-associated CHD8/kis, and microcephaly-associated ASPM/asp also showed smaller wing

186 areas and vein lengths (Fig. 4B-C). In contrast, TRPM1/Trpm within 15q13.3 and the cell

187 proliferation gene PTEN/Pten57 both showed larger wing areas and vein lengths (Fig. 4B-C).

188 Furthermore, we identified eight homologs that showed no qualitative wing phenotypes but had

189 significant changes in wing areas and vein lengths (Supp. Data 3), including CCDC101/Sgf29 in

190 distal 16p11.2, FMO5/Fmo-2, TRPM1/Trpm, DHRS11/CG9150 in 17q12, and NSUN5/Nsun5 in

191 7q11.23 (Fig. 4B-C; Supp. Data 3). These results indicate that homologs of certain CNV genes

192 may influence variations in size without causing adverse wing phenotypes, and may be

193 specifically implicated towards cellular growth mechanisms.

194

195

196

9

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

197 Homologs of CNV genes show global and tissue-specific effects during development

198 We previously showed that many of the same fly homologs of CNV genes that showed wing

199 defects in the current study also contributed towards neuronal phenotypes in the fly eye41,42,

200 suggesting a role for these genes in global development. We therefore performed ubiquitous and

201 eye-specific knockdown of fly homologs to assess tissue-specific effects in comparison to the

202 wing phenotypes. First, we used the da-GAL4 driver at 25°C to drive ubiquitous knockdown of

203 RNAi lines for 31 homologs of CNV genes, including 19 that were previously published41,42, and

204 observed complete or partial lethality at larval and pupal stages with knockdown of 10/31

205 homologs (32.3%) (Fig. 5A). Lethal phenotypes have also been documented for 43/130

206 knockout mouse models of individual CNV genes as well as for the entire deletion (Supp. Data

207 4). For example, mouse models heterozygous for the 16p11.2 deletion showed partial neonatal

208 lethality, while knockout mouse models of four individual genes within the 16p11.2 region,

209 including Ppp4C-/- and Kif22-/-, showed embryonic lethality25,58,59. In our study, the DLG1/dlg1

210 line that showed lethality with wing-specific knockdown also exhibited larval lethality with

211 ubiquitous knockdown, indicating its role in global development (Fig. 5A). In addition, six

212 homologs that showed severe wing phenotypes also showed larval or pupal lethality with

213 ubiquitous knockdown, including ALDOA/Ald and PPP4C/Pp4-19C within 16p11.2 and

214 ATXN2L/Atx2 and TUFM/mEFTu1 within distal 16p11.2 (Fig. 5A). The remaining homologs

215 that showed lethality with ubiquitous knockdown showed at least a mild qualitative or

216 quantitative wing phenotype.

217 We next compared the phenotypes observed with wing-specific knockdown of fly

218 homologs to their corresponding eye-specific knockdowns to evaluate neuronal versus non-

219 neuronal effects. To quantitatively assess the phenotypic severity of cellular defects with eye-

10

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

220 specific knockdown of fly homologs, we developed a tool called Flynotyper60 that determines the

221 degree of disorganization among the ommatidia in the adult eye. We analyzed phenotypic scores

222 obtained from Flynotyper for 66 RNAi lines of 45 fly homologs, including from previously-

223 published datasets41,42,60. We found that 37/45 homologs (82.2%) exhibited both eye and wing-

224 specific defects (Fig. 5B, Supp. Fig. 2, Supp. Data 5). Two homologs with significant eye

225 phenotypes did not show any wing phenotypes, including SPNS1/spin within distal 16p11.2 and

226 microcephaly-associated SLC25A19/Tpc161, while five homologs only showed wing-specific

227 phenotypes, including CDIPT/Pis and YPEL3/CG15309 within 16p11.2, FBXO45/Fsn and

228 OSTalpha/CG6836 within 3q29, and UQCRC2/UQCR-C2 (Fig. 5B, Supp. Fig. 2). In particular,

229 UQCRC2/UQCR-C2 showed lethality with wing-specific knockdown, suggesting potential

230 tissue-specific effects of this gene in non-neuronal cells (Fig. 5B). While most homologs

231 contributed towards both eye and wing-specific phenotypes, we observed a wide range of

232 severity in eye phenotypes that did not correlate with the severity of quantitative or qualitative

233 wing phenotypes (Fig. 5C). For example, TUFM/mEFTu1 showed a severe wing phenotype but

234 only a mild increase in eye phenotypic score, while SH2B1/Lnk, also within the distal 16p11.2

235 region, showed severe rough eye phenotypes but only a mild increase in wing size (Fig. 5D).

236 Similarly, BCL9/lgs also showed opposing tissue-specific effects with mild qualitative wing

237 phenotype and severe eye phenotype, suggesting that the role of these homologs towards

238 development differs across tissue types.

239

240 CNV genes show variable expression across different tissues in flies and humans

241 To assess how expression levels of CNV genes vary across different tissues, we first examined

242 the expression patterns of fly homologs in larval and adult tissues using the FlyAtlas Anatomical

11

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

243 Microarray dataset62. We found that 76/77 homologs with available data were expressed in at

244 least one larval and adult tissue (Supp. Fig. 3, Supp. Data 6). In general, we did not observe a

245 correlation between wing phenotype severity and expression patterns of homologs in larval or

246 adult tissues (Fig. 6A). For example, 58/77 homologs (75.3%) showed ubiquitous larval

247 expression, including both fly homologs that showed no qualitative wing phenotypes, such as

248 KCTD13/CG10465 within 16p11.2 and FBXO45/Fsn, and those with severe wing phenotypes,

249 such as PPP4C/Pp4-19C and NCBP2/Cbp20 (Fig. 6A, Supp. Fig. 3). Furthermore, 30/39

250 homologs (76.9%) that showed eye phenotypes also had ubiquitous larval expression, providing

251 further support to the observation that genes causing neuronal phenotypes may also contribute to

252 developmental phenotypes in other tissues (Supp. Data 5). Of note, 9/77 homologs (11.7%) did

253 not have any expression in the larval central nervous system, including FMO5/Fmo-2,

254 BDH1/CG8888 within 3q29, and TBX6/Doc2 within 16p11.2 (Fig. 6A, Supp. Fig. 3). However,

255 we observed wing phenotypes for 8/9 of these homologs, suggesting that they may contribute to

256 tissue-specific phenotypes outside of the nervous system. Except for the epilepsy-associated

257 SCN1A/para63, which was exclusively expressed in both the larval central nervous system (CNS)

258 and adult brain tissues, other tested neurodevelopmental genes were also expressed in non-

259 neuronal tissues (Fig. 6A).

260 We further used the GTEx Consortium dataset64 to examine tissue-specific expression of

261 150 human CNV and known neurodevelopmental genes across six tissues including brain, heart,

262 kidney, lung, liver, and muscle. We found 121 genes that were expressed in at least one adult

263 tissue, including 49 genes (32.7%) that showed ubiquitous expression across all six tissues

264 (Supp. Data 6). Of the 112 genes expressed in non-neuronal tissues, 34 did not have any

265 neuronal expression, including TBX1, FMO5 and GJA5 within 1q21.1, and ATP2A1 within distal

12

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

266 16p11.2 (Fig. 6B, Supp. Data 6). FMO5 and TBX1 also showed non-neuronal expression in

267 Drosophila tissues, suggesting that their tissue-specific expression is highly conserved (Fig. 6A).

268 Other genes showing ubiquitous expression also had preferentially high expression for specific

269 non-neuronal tissues, including ALDOA and UQCRC2 for muscle and heart for (Fig. 6B). In

270 contrast, we found nine genes that were expressed only in the adult brain, including FAM57B

271 and DOC2A within 16p11.2, as well as SCN1A, which showed similar CNS-only expression in

272 Drosophila tissues (Fig. 6B, Supp. Data 6).

273

274 Knockdown of fly homologs of CNV genes lead to disruption of basic cellular processes

275 The disruption of basic cellular processes in neuronal cells, such as cell proliferation and

276 apoptosis, have been implicated in neurodevelopmental disorders65–67. We previously identified

277 defects in cell proliferation among photoreceptors neurons in larval eye discs with knockdown of

278 16p11.2 homologs, as well as increased apoptosis with knockdown of a subset of 3q29

279 homologs41,42. Here, we explored how these basic cellular processes are altered in non-neuronal

280 cells, specifically in the developing wing disc. We targeted 27 fly homologs that showed a range

281 of adult wing phenotypes for changes in cell proliferation and apoptosis, using anti-phospho-

282 H3 Ser10 (pH3) and anti-Drosophila caspase-1 (dcp1), respectively, in the third instar

283 larval wing discs. We identified 23/27 homologs that showed significant increases in apoptotic

284 cells compared to controls, including seven homologs, such as PPP4C/Pp4-19C, ATXN2L/Atx2,

285 and AATF/Aatf , which showed dcp1 staining across the entire larval wing pouch (Fig. 7A-B,

286 Supp. Figs. 4-5, Supp. Data 7). In addition, 16/27 genes showed decreased levels of

287 proliferation, including eight homologs which also showed apoptosis defects, such as

288 CYFIP1/Sra-1 within 15q11.2, SH2B1/Lnk, and the microcephaly gene KIF11/Klp61F (Fig. 7A

13

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

289 and 7C, Supp. Figs. 4-5, Supp. Data 7). All six of the tested homologs with severe adult wing

290 phenotypes showed both increased apoptosis and decreased proliferation (Supp. Data 7).

291 Similarly, 3/4 homologs of genes showing lethality with wing-specific knockdown also showed

292 defects in apoptosis or proliferation, with the exception of ACACA/ACC (Supp. Figure 4, Supp.

293 Data 7). As bxMS1096-GAL4 is located on the X-, we expected to see more severe

294 defects in males compared with females with knockdown of homologs due to the X-linked

295 dosage compensation54,55. However, knockdown of 3/11 tested homologs with sex-specific

296 differences in adult wing phenotypes, including BCL9/lgs, CYFIP1/Sra-1, and

297 DNAJC30/CG11035 within 7q11.23, showed significantly decreased levels of cell proliferation

298 in females but no change for males compared to their respective controls, suggesting a sex-

299 specific effect of these genes for cell proliferation (Supp. Fig. 5, Supp. Data 7). Overall, our

300 results suggest that cell proliferation and apoptosis play an important role towards development

301 in both neuronal and non-neuronal tissues.

302

303 Homologs of candidate CNV genes disrupt conserved signaling pathways

304 Several conserved signaling pathways that are active in a spatial and temporal manner in the

305 larval wing disc, such as Wnt, Hedgehog, BMP, and Notch signaling, regulate the anterior-

306 posterior (A/P) and dorsal-ventral (D/V) boundaries to determine accurate morphology and vein

307 patterning in the adult wing48,49,68–70. For example, Wnt and Notch signaling pathways both act

308 along the D/V boundary to determine cell fate71,72, while Hedgehog signaling is dependent upon

309 expression of both engrailed in the posterior compartment and patched along the A/P border73,74.

310 Furthermore, O’Roak and colleagues showed that genes identified from de novo mutations in

311 patients with autism are linked to β-catenin/Wnt pathway75. In addition, familial loss-of-function

14

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

312 mutations in the human hedgehog signaling pathway gene PTCH1 are implicated in basal cell

313 nevus syndrome, leading to basal cell carcinoma76,77.

314 Based on adult wing phenotypes and disruptions to cellular processes, we next tested

315 whether knockdown of 14 fly homologs disrupt conserved signaling pathways in the third instar

316 larval wing disc (Supp. Data 7). In particular, we evaluated the role of Wnt, Hedgehog, and

317 Notch signaling pathways by testing the expression patterns of four key within these

318 pathways, including wingless (Wnt), patched (Hedgehog), engrailed (Hedgehog), and delta

319 (Notch). We found that 9/14 homologs, including 8/10 homologs showing severe wing

320 phenotypes or lethality, exhibited disruptions in at least one signaling pathway. For example, five

321 homologs with severe or lethal phenotypes showed disruptions of all four signaling pathways,

322 including AATF/Aatf, NCBP2/Cbp20, POLR3E/Sin, PPP4C/Pp4-19C, and KIF11/Klp61F (Fig.

323 8, Supp. Data 7). Our observations are in concordance with previous findings by Swarup and

324 colleagues, who showed that PPP4C/Pp4-19C is a candidate regulator of Wnt and Notch

325 signaling pathways in Drosophila larval wing discs78. Furthermore, two genes from the 3q29

326 region, DLG1/dlg1 and MFI2/Tsf2, showed altered expression patterns for delta and patched but

327 not for engrailed, indicating that they selectively interact with the Hedgehog as well as Notch

328 signaling pathway (Supp. Fig. 6). In fact, Six and colleagues showed that Dlg1 directly binds to

329 the PDZ-binding domain of Delta179. In contrast, ACACA/ACC and UQCRC2/UQCR-C2 showed

330 no changes in expression patterns for any of the four signaling proteins tested, suggesting that the

331 observed lethality could be due to other cellular mechanisms (Supp. Fig. 6). We conclude that a

332 subset of homologs disrupt the expression of key proteins in signaling pathways, potentially

333 accounting for the developmental phenotypes observed in the adult wings.

334

15

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

335 Connectivity patterns of candidate genes vary across human tissue-specific networks

336 We examined patterns of connectivity for the nine candidate genes which showed disruptions of

337 signaling pathways within the context of human brain, heart, and kidney-specific gene

338 interaction networks80. These tissue-specific networks were constructed using Bayesian

339 classifier-generated probabilities for pairwise genetic interactions based on co-expression data80.

340 We calculated the lengths of the shortest paths between each candidate gene and 267 Wnt,

341 Notch, and Hedgehog pathway genes in each network as a proxy for connectivity (Supp. Data

342 8). In all three networks, each of the candidate genes were connected to a majority of the tested

343 signaling pathway genes, suggesting that our results have translational relevance towards human

344 developmental pathways (Fig. 9A, Supp. Fig. 7). Interestingly, we observed a higher

345 connectivity (i.e. shorter path distances) between candidate genes and Wnt and Hedgehog

346 pathway genes in the brain-specific network compared to the heart and kidney-specific networks

347 (Fig. 9B). We further identified enrichments for genes involved in specific biological processes

348 among the connector genes that were located in the shortest paths within neuronal and non-

349 neuronal tissue-specific networks (Fig. 9C, Supp. Data 8). For example, axon-dendrite

350 transport, dopaminergic signaling, and signal transduction functions were enriched among

351 connector genes only for the brain-specific network, while organelle organization and

352 ubiquitination were enriched among connector genes only for kidney and heart networks (Fig.

353 9C). However, several core biological processes, such as cell cycle, protein metabolism,

354 transcriptional regulation, and RNA processing/splicing, were enriched among connector genes

355 within all three tissue-specific networks (Fig. 9C). Our analysis highlights that human CNV

356 genes potentially interact with developmental signaling pathways in an ubiquitous manner, but

357 may affect different biological processes in neuronal and non-neuronal tissues.

16

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

358 DISCUSSION

359 We used the Drosophila wing as a model to identify key CNV genes involved in non-neuronal

360 phenotypes associated with CNV disorders. We tested fly homologs of 79 genes and identified

361 multiple homologs within each CNV region that exhibited strong phenotypes indicative of

362 developmental disruptions. Several themes have emerged from our study highlighting the

363 importance of fly homologs of CNV genes towards both global and tissue-specific phenotypes

364 associated with human CNV disorders.

365 First, we found that homologs of CNV genes contribute towards developmental

366 phenotypes through ubiquitous roles in neuronal and non-neuronal tissues. Although we did not

367 study models for the entire CNV, nearly all individual fly homologs of CNV genes contribute to

368 wing-specific developmental phenotypes. It is likely that these genes may also contribute to

369 additional phenotypes in other tissues that we did not assess. In fact, a subset of these genes also

370 showed early lethality with ubiquitous knockdown in addition to severe or lethal wing-specific

371 phenotypes. However, we found no correlation between the severity of the eye and wing

372 phenotypes, suggesting tissue-specific effects of these homologs towards developmental

373 phenotypes. In contrast, fly homologs of known neurodevelopmental genes generally showed

374 milder wing phenotypes compared with eye phenotypes, indicating a more neuronal role for

375 these genes. While our study only examined a subset of CNV genes with Drosophila homologs,

376 phenotypic data from knockout mouse models also support a global developmental role for

377 individual CNV genes. In fact, 44/130 knockout models of CNV genes within the Mouse

378 Genome Informatics (MGI) database81 exhibited non-neuronal phenotypes, including 20

379 homologs of CNV genes that showed both neuronal and non-neuronal phenotypes (Supp. Data

380 4). For example, knockout mouse models of Dlg1-/- show defects in dendritic growth and

17

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

381 branching in the developing nervous system, in addition to craniofacial features and multiple

382 kidney and urinary tract defects82–85. Furthermore, Chapman and colleagues showed that

383 knockout of Tbx6-/- caused defects in mesodermal and neuronal differentiation early in

384 development, leading to abnormal vascular, tail bud, and neural tube morphology86. These

385 observations further support our findings that most fly homologs of CNV genes have a global

386 role in development that could account for the observed non-neuronal phenotypes.

387 Second, based on tissue-specific phenotypes, we identified fly homologs of CNV genes

388 that are key regulators of conserved cellular processes important for development. For example,

389 9/10 homologs with severe or lethal adult wing phenotypes also exhibited defects in cell

390 proliferation and apoptosis during development. In fact, we found concordance between cellular

391 processes affected by wing and eye-specific knockdown of homologs of genes within 16p11.2

392 and 3q29 regions, including decreased proliferation for MAPK3/rl and increased apoptosis for

393 NCBP2/Cbp20 and DLG1/dlg141,42. While eye-specific knockdown of BDH1/CG8888 showed

394 decreased cell proliferation in larval eye discs42, we found increased cell proliferation with wing-

395 specific knockdown, suggesting a tissue-specific effect for this gene. Notably, at least one fly

396 homolog per CNV region showed defects in cell proliferation or apoptosis, suggesting that these

397 conserved cellular processes may be relevant to CNV pathogenicity. For example, ATXN2L/Atx2,

398 SH2B1/Lnk, and CCDC101/Sgf29 each showed decreased proliferation and increased apoptosis,

399 suggesting an underlying common conserved cellular mechanism for the distal 16p11.2 deletion.

400 Furthermore, a subset of these genes also disrupted multiple signaling pathways, indicating a

401 potential role for these genes as key regulators of developmental processes. We specifically

402 identified five genes whose knockdown caused disruptions of Wnt, Notch, and hedgehog

403 signaling pathways. Each of these genes have important roles in cell cycle regulation, apoptosis,

18

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

404 transcription, or RNA processing, based on annotations87,88. In fact, we found

405 that the RNA transport protein NCBP2/Cbp2089, which we recently identified as a key modifier

406 gene for the 3q29 deletion42, interfaced with all three signaling pathways. Furthermore, AATF

407 disrupts apoptosis and promotes cell cycle progression through displacement of HDAC190–92,

408 while PPP4C promotes spindle organization at the centromeres during mitosis93. While we only

409 evaluated the role of these genes towards development in a single fly tissue, our additional

410 analysis of human gene interaction networks showed strong connectivity between the CNV

411 genes and signaling pathways in multiple neuronal and non-neuronal human tissues. In fact, cell

412 cycle genes were enriched among the connector genes in all three tissue-specific networks,

413 further emphasizing the role of cell cycle processes towards developmental phenotypes. Notably,

414 we also observed certain biological processes enriched among connector genes that were specific

415 to neuronal or non-neuronal tissues, indicating that haploinsufficiency of genes within CNV

416 regions may disrupt different biological processes in a tissue-specific manner.

417 Overall, we show that fly homologs of most CNV genes contribute towards global

418 developmental phenotypes, although exactly how they contribute toward such phenotypes varies

419 between neuronal and non-neuronal tissues. Previous functional studies for CNV disorders have

420 focused primarily on identifying candidate genes for the observed neuronal phenotypes. In this

421 study, we identified several homologs of CNV genes that are responsible for non-neuronal

422 phenotypes, as well as novel associations between these genes and conserved biological

423 processes and pathways. We therefore propose that multiple genes within each CNV disrupt

424 global and tissue-specific processes during development and contribute to the wide range of non-

425 neuronal phenotypes associated with CNV disorders (Fig. 10). This multigenic model for non-

426 neuronal phenotypes in CNV disorders is in line with our previous model for neuronal

19

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

427 phenotypes of these disorders, as opposed to models where individual causative genes are

428 responsible for specific phenotypes41,42,94. Our study further exemplifies the utility of evaluating

429 non-neuronal phenotypes in addition to neuronal phenotypes in models of individual genes and

430 CNV regions associated with developmental disorders, including future studies in mammalian or

431 cellular model systems. Further studies exploring how CNV genes interact with each other and

432 with other developmental pathways could more fully explain the conserved mechanisms

433 underlying global developmental defects and identify potential therapeutic targets for these

434 disorders.

20

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

435 METHODS

436 Fly stocks and genetics

437 We tested 59 Drosophila homologs for 130 human genes that span across 10 pathogenic CNV

438 regions associated with neurodevelopmental disorders (1q21.1, 3q29, 7q11.23, 15q11.2, 15q13.3,

439 16p11.2, distal 16p11.2, 16p12.1, 16p13.11, and 17q12)22 (Supp. Data 1). In addition, we

440 evaluated fly homologs of 20 human genes known to be in involved in neurodevelopmental

441 disorders60,95 (Supp. Data 1). These include genes involved in beta-catenin signaling pathway (5

442 genes), core genes implicated in neurodevelopmental disorders (8 genes), and genes associated

443 with microcephaly (7 genes)96. We used the DRSC Integrative Ortholog Prediction Tool

444 (DIOPT, v.7.1) to identify the fly homologs for each human gene53 (Supp. Data 1).

445 To knockdown individual genes in specific tissues, we used RNA interference (RNAi)

446 and the UAS-GAL4 system (Fig. 2A), a well-established tool that allows for tissue-specific

447 expression of a gene of interest97. RNAi lines were obtained from Vienna Drosophila Resource

448 Center (VDRC) that include both GD and KK lines. We tested a total of 136 lines in our final

449 data analysis (Supp. Data 9), after eliminating KK lines with additional insertion that drives the

450 overexpression of the Tiptop (tio) transcription factor98,99. A complete list of stock numbers and

451 full genotypes for all RNAi lines used in this study is presented in Supp. Data 9. We used the

452 bxMS1096-GAL4/FM7c;;UAS-Dicer2/TM6B driver for wing-specific knockdown and w1118;GMR-

453 GAL4;UAS-Dicer2 driver (Claire Thomas, Penn State University) for eye-specific knockdown of

454 RNAi lines. Ubiquitous knockdown experiments were performed using the w;da-GAL4;+ driver

455 (Scott Selleck, Penn State University). For all experiments, we used appropriate GD (w1118,

456 VDRC# 60000) or KK (y,w1118; P{attP,y+,w3`}, VDRC# 60100) lines as controls to compare

457 against lines with knockdown of individual homologs. All fly lines were reared on standard yeast

21

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

458 Drosophila medium at room temperature. All crosses were set and maintained at 25°C, except

459 for the eye knockdown experiments which were maintained at 30°C.

460

461 Phenotypic analysis of adult wing images

462 Adult progeny were isolated from crosses between RNAi lines and bxMS1096-GAL4 driver shortly

463 after eclosion, and kept at 25°C until day 2-5 (Fig. 2A). At that point, the progeny were frozen at

464 -80°C, and were then moved to -20°C prior to imaging and storage. Approximately 20-25

465 progeny, both male and female, were collected for each RNAi line tested. The adult wings were

466 plucked from frozen flies and mounted on a glass slide. The slides were covered with a coverslip

467 and sealed using clear nail polish. Adult wing images were captured using a Zeiss Discovery

468 V20 stereoscope (Zeiss, Thornwood, NY, USA), with a ProgRes Speed XT Core 3 camera and

469 CapturePro v.2.8.8 software (Jenoptik AG, Jena, Germany) at 40X magnification.

470 For each non-lethal RNAi line, we scored the adult wing images for five qualitative

471 phenotypes, including wrinkled wing, discoloration, missing veins, ectopic veins, and bristle

472 planar polarity defects, on a scale of 1 (no phenotype) to 5 (lethal) (Fig. 3C). Lines showing

473 severely wrinkled wings or lethality were scored as 4 (severe) or 5 (lethal) for all five

474 phenotypes. We calculated the frequency of each phenotypic score (i.e. mild bristle polarity,

475 moderate discoloration) across all of the wing images for each line (Fig. 3A-B), and then

476 performed k-means clustering of these values to generate five clusters for overall wing

477 phenotypes (Fig. 2C). For quantitative analysis of wing phenotypes, we used the Fiji ImageJ

478 software100 to calculate the wing area using the Measure Area tool, and calculated the lengths of

479 longitudinal veins L2, L3, L4, and L5 as well as the anterior and posterior crossveins (ACV and

480 PCV), by tracing individual veins using the Segmented Line tool (Fig. 4A, Supp. Data 2). We

22

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

481 determined discordant homologs when RNAi lines for the same homologs showed inconsistent

482 wing phenotypes. For each homolog with multiple RNAi lines, we checked discordance among

483 RNAi lines for no phenotype versus any qualitative or quantitative phenotypes, followed by

484 discordance for small or large wing measurement phenotypes (Supp. Data 3).

485

486 Phenotypic analysis of adult eye images

487 We crossed RNAi lines with GMR-GAL4 to achieve eye-specific knockdown of homologs of

488 CNV and known neurodevelopmental genes. Adult 2-3-day old female progenies from the

489 crosses were collected, immobilized by freezing at -80°C, and then moved to -20°C prior to

490 imaging and storage. Flies were mounted on Blu-tac (Bostik Inc, Wauwatosa, WI, USA) and

491 imaged using an Olympus BX53 compound microscope with LMPLan N 20X air objective using

492 a DP73 c-mount camera at 0.5X magnification (Olympus Corporation, Tokyo, Japan). CellSens

493 Dimension software (Olympus Corporation, Tokyo, Japan) was used to capture the eye images,

494 which were then stacked using the Zerene Stacker software (Zerene Systems LLC, Richland,

495 WA, USA). All eye images presented in this study are maximum projections of 20 consecutive

496 optical z-sections, at a z-step size of 12.1μm. Finally. we used our computational method called

497 Flynotyper (https://flynotyper.sourceforge.net) to quantify the degree of rough eye phenotypes

498 present due to knockdown of homologs of CNV or neurodevelopmental genes60. Flynotyper

499 scores for homologs of 16p11.2 and 3q29, as well as select core neurodevelopmental genes, were

500 derived from our previous studies41,42,60.

501

502

503

23

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

504 Immunohistochemistry

505 Wing imaginal discs from third instar larvae were dissected in 1X PBS. The tissues were fixed

506 using 4% paraformaldehyde and blocked using 1% bovine serum albumin (BSA). The wing discs

507 were incubated with primary antibodies using appropriate dilutions overnight at 4°C. We used

508 the following primary antibodies: mouse monoclonal anti-pHistone3 (S10) (1:100 dilutions, Cell

509 Signaling 9706L), rabbit polyclonal anti-cleaved Drosophila Dcp1 (Asp216) (1:100 dilutions,

510 Cell Signaling 9578S), mouse monoclonal anti-Wingless (1:200 dilutions, DSHB, 4D4), mouse

511 monoclonal anti-Patched (1:50 dilutions, DSHB, Drosophila Ptc/APA1), mouse monoclonal

512 anti-Engrailed (1:50 dilutions, DSHB, 4D9), and mouse monoclonal anti-Delta (1:50 dilutions,

513 DSHB, C594.9B). Following incubation with primary antibodies, the wing discs were washed

514 and incubated with secondary antibodies at 1:200 dilution for two hours at room temperature.

515 We used the following secondary antibodies: Alexa Fluor 647 dye goat anti-mouse (A21235,

516 Molecular Probes by Invitrogen/Life Technologies), Alexa Fluor 568 dye goat anti-rabbit

517 (A11036, Molecular Probes by Invitrogen/Life Technologies), and Alexa Fluor 568 dye goat

518 anti-mouse (A11031, Molecular Probes by Invitrogen/Life Technologies). All washes and

519 antibody dilutions were made using 0.3% PBS with Triton-X.

520 Third instar larvae wing imaginal discs were mounted in Prolong Gold antifade reagent

521 with DAPI (Thermo Fisher Scientific, P36930) for imaging using an Olympus Fluoview FV1000

522 laser scanning confocal microscope (Olympus America, Lake Success, NY). Images were

523 acquired using FV10-ASW 2.1 software (Olympus, Waltham, MA, USA). Composite z-stack

524 images were analyzed using the Fiji ImageJ software100. To calculate the number of pH3 positive

525 cells within the wing pouch area of the wing discs, we used the AnalyzeParticles function in

526 ImageJ, while manual counting was used to quantify Dcp1 positive cells. We note that cell

24

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

527 proliferation and apoptosis staining for NCBP2/Cbp20, DLG1/dlg1, BDH1/CG8888, and

528 FBXO45/Fsn were previously published42.

529

530 Statistical analysis

531 Significance for the wing area and vein length measurements, cell counts for proliferation and

532 apoptosis, and Flynotyper scores were compared to appropriate GD or KK controls using one-

533 tailed or two-tailed Mann-Whitney tests. P-values for each set of experiments were corrected for

534 multiple testing using Benjamini-Hochberg correction. All statistical and clustering analysis was

535 performed using R v.3.6.1 (R Center for Statistical Computing, Vienna, Austria). Details for the

536 statistical tests performed for each dataset are provided in Supp. Data 10.

537

538 Expression data analysis

539 We obtained tissue-specific expression data for fly homologs of CNV genes from the FlyAtlas

540 Anatomical Microarray dataset62. Raw FPKM (fragments per kilobase of transcript per million

541 reads) expression values for each tissue were categorized as follows: <10, no expression; 10-100,

542 low expression; 100-500, moderate expression; 500-1000, high expression; and >1000, very high

543 expression (Supp. Data 6). The median expression among midgut, hindgut, Malpighian tube,

544 and (for adult only) crop tissues was used to represent the overall gut expression. We similarly

545 obtained human tissue-specific expression data for CNV genes from the GTEx Consortium v.1.2

546 RNA-Seq datasets64. Median TPM (transcripts per million reads) expression values for each

547 tissue were categorized as follows: <3, no expression; 3-10, low expression; 10-25, moderate

548 expression; 25-100, high expression; and >100, very high expression (Supp. Data 6). The

549 median expression among all brain and heart sub-tissues was used to represent brain and heart

25

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

550 expression, while the median expression among all colon, esophagus, small intestine, and

551 stomach sub-tissues was used to represent digestive tract expression. Preferential gene

552 expression for a particular tissue within the GTEx dataset was determined if the expression

553 values for that tissue were greater than the third quartile of all tissue expression values for that

554 gene, plus 1.5 times the interquartile range. Venn diagrams were generated using the Venny

555 webtool (http://bioinfogp.cnb.csic.es/tools/venny) (Supp. Fig. 3).

556

557 Network analysis

558 We obtained human tissue-specific gene interaction networks for brain, heart, and kidney tissues

559 from the GIANT network database80 within HumanBase (https://hb.flatironinstitute.org). These

560 networks were built by training a Bayesian classifier based on tissue-specific gene co-expression

561 datasets, which then assigned a posterior probability for interactions between each pair of genes

562 within the genome for a particular tissue. We downloaded the “Top edge” version of each tissue-

563 specific network, and extracted all gene pairs with posterior probabilities >0.2 to create sub-

564 networks containing the top ~0.5% tissue-specific interactions. Next, we identified the shortest

565 paths in each sub-network between human CNV genes whose fly homologs disrupted signaling

566 pathways in the larval wing disc and human genes within each disrupted pathway, using the

567 inverse of the posterior probability as weights for each edge in the network. Gene sets from the

568 human Notch (KEGG:map04330), Wnt (KEGG:map04310) and Hedgehog pathways

569 (KEGG:map04340) were curated from the Kyoto Encyclopedia of Genes and Genomes (KEGG)

570 pathway database101. Using the NetworkX Python package102, we calculated the shortest distance

571 between each CNV gene and pathway gene, and identified connecting genes that were within

572 each of the shortest paths for the three tissue-specific networks. We further tested for enrichment

26

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

573 of Gene Ontology (GO) terms (PantherDB GO-Slim) among the connector genes using the

574 PantherDB Gene List Analysis tool103. Lists of the shortest paths and connector genes in each

575 tissue-specific network, as well as enriched GO terms for sets of connector genes, are provided

576 in Supp. Data 8. Gene networks were visualized using Cytoscape v.3.7.2104 using an edge-

577 weighted spring embedded layout.

578

579 Mouse and human phenotypic data analysis

580 Phenotypic data for mouse models of CNV gene homologs, categorized using top-level

581 Mammalian Phenotype Ontology terms, were obtained from the Mouse Genome Informatics

582 (MGI) database81 (Supp. Data 4). Phenotypic data for human carriers of pathogenic CNVs were

583 obtained from the DECIPHER public database9. Clinical phenotypes for each CNV carrier were

584 categorized by top-level Human Phenotype Ontology terms105 using the Orange3 Bioinformatics

585 software library (https://orange-bioinformatics.readthedocs.io), and the frequency of individuals

586 carrying each top-level phenotype term was calculated for each of the ten tested pathogenic

587 CNVs.

588

27

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

589 DATA AVAILABILITY

590 All data supporting the findings of this study are available within the paper and its

591 supplementary information files.

592

593 CODE AVAILABILITY

594 All source code for data analysis in this manuscript, including scripts for k-means clustering of

595 fly phenotypes, network connectivity of CNV and developmental pathway genes, and extraction

596 of top-level human phenotype terms, are available on the Girirajan lab GitHub page at

597 https://github.com/girirajanlab/CNV_wing_project.

598

599 ACKNOWLEDGEMENTS

600 The authors thank members of the Girirajan Lab for their helpful discussions and comments on

601 the manuscript. This study makes use of data generated by the DECIPHER community. A full

602 list of centers who contributed to the generation of the data is available from

603 http://decipher.sanger.ac.uk and via email from [email protected]. Funding for the project

604 was provided by the Wellcome Trust. This work was supported by NIH R01-GM121907 and

605 resources from the Huck Institutes of the Life Sciences to S.G., and NIH T32-GM102057 to M.J.

606

607 CONTRIBUTIONS

608 T.Y., M.J., S.Y., and S.G. designed the study. T.Y., S.Y., L.P., S.K., D.J.G., A.S., Y.M., J.I., and

609 Z.C.L. performed the functional experiments. T.Y. and M.J. performed the expression and

610 network experiments. T.Y., M.J., and S.G. analyzed the data and wrote the manuscript with input

611 from all authors.

28

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

612 COMPETING OF INTERESTS

613 The authors declare that they have no competing interests.

29

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

614 REFERENCES 615 616 1. Girirajan, S., Campbell, C. D. & Eichler, E. E. Human Copy Number Variation and 617 Complex Genetic Disease. Annu. Rev. Genet. 45, 203–226 (2011). 618 2. Malhotra, D. & Sebat, J. CNVs: harbingers of a rare variant revolution in psychiatric 619 genetics. Cell 148, 1223–1241 (2012). 620 3. Cooper, G. M. et al. A copy number variation morbidity map of developmental delay. Nat. 621 Genet. 43, 838–846 (2011). 622 4. Zhang, F., Gu, W., Hurles, M. E. & Lupski, J. R. Copy Number Variation in Human 623 Health, Disease, and Evolution. Annu. Rev. Genomics Hum. Genet. 10, 451–481 (2009). 624 5. Greenway, S. C. et al. De novo copy number variants identify new genes and loci in 625 isolated sporadic tetralogy of Fallot. Nat. Genet. 41, 931–935 (2009). 626 6. Glessner, J.T. et al. Increased Frequency of De Novo Copy Number Variations in 627 Congenital Heart Disease by Integrative Analysis of SNP Array and Exome Sequence 628 Data. Circ Res. 115, 884–896 (2014). 629 7. Sanna-Cherchi, S. et al. Copy-number disorders are a common cause of congenital kidney 630 malformations. Am. J. Hum. Genet. 91, 987–997 (2012). 631 8. Zahnleiter, D. et al. Rare copy number variants are a common cause of short stature. PLoS 632 Genet. 9, e1003365 (2013). 633 9. Firth, H. V. et al. DECIPHER: Database of Chromosomal Imbalance and Phenotype in 634 Humans Using Ensembl Resources. Am. J. Hum. Genet. 84, 524–533 (2009). 635 10. Mefford, H. C. et al. Recurrent rearrangements of chromosome 1q21.1 and variable 636 pediatric phenotypes. N. Engl. J. Med. 359, 1685–99 (2008). 637 11. Brunetti-Pierri, N. et al. Recurrent reciprocal 1q21.1 deletions and duplications associated 638 with microcephaly or macrocephaly and developmental and behavioral abnormalities. Nat. 639 Genet. 40, 1466–1471 (2008). 640 12. Christiansen, J. et al. Chromosome 1q21.1 contiguous gene deletion is associated with 641 congenital heart disease. Circ. Res. 94, 1429–35 (2004). 642 13. Pober, B. R. Williams-Beuren syndrome. N. Engl. J. Med. 362, 239–52 (2010). 643 14. Ewart, A. K. et al. A human vascular disorder, supravalvular aortic stenosis, maps to 644 . Proc. Natl. Acad. Sci. U. S. A. 90, 3226–30 (1993). 645 15. Romeo, S. et al. Search for genetic variants of the SYNTAXIN 1A (STX1A) gene: The - 646 352 A>T variant in the STX1A promoter associates with impaired glucose metabolism in 647 an Italian obese population. Int. J. Obes. 32, 413–420 (2008). 648 16. Frangiskakis, J. M. et al. LIM-kinase1 hemizygosity implicated in impaired visuospatial 649 constructive cognition. Cell 86, 59–69 (1996). 650 17. Tassabehji, M. et al. GTF2IRD1 in craniofacial development of humans mice. Science 651 310, 1184–1187 (2005). 652 18. Lindsay, E. A. et al. Tbx1 haploinsufficiency in the DiGeorge syndrome region causes 653 aortic arch defects in mice. 410, 97–101 (2001). 654 19. Mefford, H. C. et al. Recurrent reciprocal genomic rearrangements of 17q12 are 655 associated with renal disease, diabetes, and epilepsy. Am. J. Hum. Genet. 81, 1057–1069 656 (2007). 657 20. Clissold, R. L. et al. Chromosome 17q12 microdeletions but not intragenic HNF1B 658 mutations link developmental kidney disease and psychiatric disorder. Kidney Int. 90, 659 203–11 (2016).

30

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

660 21. Weiss, L. A. et al. Association between Microdeletion and Microduplication at 16p11.2 661 and Autism. N. Engl. J. Med. 358, 667–675 (2008). 662 22. Girirajan, S. et al. Phenotypic Heterogeneity of Genomic Disorders and Rare Copy- 663 Number Variants. N. Engl. J. Med. 367, 1321–1331 (2012). 664 23. Bernier, R. et al. Disruptive CHD8 Mutations Define a Subtype of Autism Early in 665 Development. Cell 158, 263–276 (2014). 666 24. Ostergaard, P. et al. Mutations in KIF11 cause autosomal-dominant microcephaly variably 667 associated with congenital lymphedema and chorioretinopathy. Am. J. Hum. Genet. 90, 668 356–62 (2012). 669 25. Horev, G. et al. Dosage-dependent phenotypes in models of 16p11.2 lesions found in 670 autism. Proc. Natl. Acad. Sci. 108, 17076–17081 (2011). 671 26. Pucilowska, J. et al. The 16p11.2 Deletion Mouse Model of Autism Exhibits Altered 672 Cortical Progenitor Proliferation and Brain Cytoarchitecture Linked to the ERK MAPK 673 Pathway. J. Neurosci. 35, 3190–3200 (2015). 674 27. Portmann, T. et al. Behavioral abnormalities and circuit defects in the basal ganglia of a 675 mouse model of 16p11.2 deletion syndrome. Cell Rep. 7, 1077–1092 (2014). 676 28. Rutkowski, T. P. et al. Behavioral changes and growth deficits in a CRISPR engineered 677 mouse model of the schizophrenia-associated 3q29 deletion. Mol. Psychiatry (2019) 678 doi:10.1038/s41380-019-0413-5. 679 29. Baba, M. et al. Psychiatric-disorder-related behavioral phenotypes and cortical 680 hyperactivity in a mouse model of 3q29 deletion syndrome. Neuropsychopharmacology 681 44, 2125–2135 (2019). 682 30. Arbogast, T. et al. Reciprocal Effects on Neurocognitive and Metabolic Phenotypes in 683 Mouse Models of 16p11.2 Deletion and Duplication Syndromes. PLoS Genet. 12, 684 e1005709 (2016). 685 31. Haller, M., Au, J., O’Neill, M. & Lamb, D. J. 16p11.2 MAZ is a 686 dosage-sensitive regulator of genitourinary development. Proc. Natl. Acad. Sci. 115, 687 E1849–E1858 (2018). 688 32. Yadav, S. et al. TAOK2 Kinase Mediates PSD95 Stability and Dendritic Spine Maturation 689 through Septin7 Phosphorylation. Neuron 93, 379–393 (2017). 690 33. Richter, M. et al. Altered TAOK2 activity causes autism-related neurodevelopmental and 691 cognitive abnormalities through RhoA signaling. Mol. Psychiatry 24, 1329–1350 (2019). 692 34. Golzio, C. et al. KCTD13 is a major driver of mirrored neuroanatomical phenotypes of the 693 16p11.2 copy number variant. Nature 485, 363–367 (2012). 694 35. Escamilla, C. O. et al. Kctd13 deletion reduces synaptic transmission via increased RhoA. 695 Nature 551, 227–231 (2017). 696 36. Ip, J. P. K. et al. Major vault protein, a candidate gene in 16p11.2 microdeletion 697 syndrome, is required for the homeostatic regulation of visual cortical plasticity. J. 698 Neurosci. 38, 2017–2034 (2018). 699 37. Dickinson, M. E. et al. High-throughput discovery of novel developmental phenotypes. 700 Nature 537, 508–514 (2016). 701 38. Wangler, M. F., Yamamoto, S. & Bellen, H. J. Fruit flies in biomedical research. Genetics 702 199, 639–653 (2015). 703 39. Chien, S. Homophila: human disease gene cognates in Drosophila. Nucleic Acids Res. 30, 704 149–151 (2002). 705 40. Reiter, L. T., Potocki, L., Chien, S., Gribskov, M. & Bier, E. A systematic analysis of

31

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

706 human disease-associated gene sequences in Drosophila melanogaster. Genome Res. 11, 707 1114–1125 (2001). 708 41. Iyer, J. et al. Pervasive genetic interactions modulate neurodevelopmental defects of the 709 autism-associated 16p11.2 deletion in Drosophila melanogaster. Nat. Commun. 9, 2548 710 (2018). 711 42. Singh, M. D. et al. NCBP2 modulates neurodevelopmental defects of the 3q29 deletion in 712 Drosophila and X. laevis models. bioRxiv 614750 (2019) doi:10.1101/614750. 713 43. Molnar, C. et al. Signalling Pathways in Development and Human Disease: A Drosophila 714 Wing Perspective. in Human Genetic Diseases (InTech, 2011). doi:10.5772/23858. 715 44. Dworkin, I. & Gibson, G. Epidermal growth factor receptor and transforming growth 716 factor-β signaling contributes to variation for wing shape in Drosophila melanogaster. 717 Genetics 173, 1417–1431 (2006). 718 45. Testa, N. D. & Dworkin, I. The sex-limited effects of mutations in the EGFR and TGF-β 719 signaling pathways on shape and size sexual dimorphism and allometry in the Drosophila 720 wing. Dev. Genes Evol. 226, 159–171 (2016). 721 46. Yan, S. J., Gu, Y., Li, W. X. & Fleming, R. J. Multiple signaling pathways and a selector 722 protein sequentially regulate Drosophila wing development. Development 131, 285–298 723 (2004). 724 47. Strigini, M. & Cohen, S. M. A Hedgehog activity gradient contributes to AP axial 725 patterning of the Drosophila wing. Development 124, 4697–4705 (1997). 726 48. Diaz de la Loza, M. C. & Thompson, B. J. Forces shaping the Drosophila wing. Mech. 727 Dev. 144, 23–32 (2017). 728 49. Bier, E. Drosophila, the golden bug, emerges as a tool for human genetics. Nat. Rev. 729 Genet. 6, 9–23 (2005). 730 50. Wu, Y. et al. A Drosophila model for Angelman syndrome. Proc. Natl. Acad. Sci. 105, 731 12399–12404 (2008). 732 51. Yamamoto, S. et al. A Drosophila genetic resource of mutants to study mechanisms 733 underlying human genetic diseases. Cell 159, 200–214 (2014). 734 52. Kochinke, K. et al. Systematic Phenomics Analysis Deconvolutes Genes Mutated in 735 Intellectual Disability into Biologically Coherent Modules. Am. J. Hum. Genet. 98, 149– 736 164 (2016). 737 53. Hu, Y. et al. An integrative approach to ortholog prediction for disease-focused and other 738 functional studies. BMC Bioinformatics 12, 357 (2011). 739 54. Capdevila, J. & Guerrero, I. Targeted expression of the signaling molecule 740 induces pattern duplications and growth alterations in Drosophila wings. 741 EMBO J. 13, 4459–4468 (1994). 742 55. Lindström, R., Lindholm, P., Palgi, M., Saarma, M. & Heino, T. I. In vivo screening 743 reveals interactions between Drosophila Manf and genes involved in the mitochondria and 744 the ubiquinone synthesis pathway. BMC Genet. 18, 52 (2017). 745 56. Nielsen, J. et al. A mouse model of the schizophrenia-associated 1q21.1 microdeletion 746 syndrome exhibits altered mesolimbic dopamine transmission. Transl. Psychiatry 7, 1261 747 (2017). 748 57. Huang, H. et al. PTEN affects cell size, cell proliferation and apoptosis during Drosophila 749 eye development. Development 126, 5365–5372 (1999). 750 58. Toyo-oka, K. et al. Protein phosphatase 4 catalytic subunit regulates Cdk1 activity and 751 microtubule organization via NDEL1 dephosphorylation. J. Cell Biol. 180, 1133–1147

32

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

752 (2008). 753 59. Ohsugi, M. et al. Kid-Mediated Chromosome Compaction Ensures Proper Nuclear 754 Envelope Formation. Cell 132, 771–782 (2008). 755 60. Iyer, J. et al. Quantitative assessment of eye phenotypes for functional genetic studies 756 using Drosophila melanogaster. G3 Genes, Genomes, Genet. 6, 1427–1437 (2016). 757 61. Rosenberg, M. J. et al. Mutant deoxynucleotide carrier is associated with congenital 758 microcephaly. Nat. Genet. 32, 175–179 (2002). 759 62. Chintapalli, V. R., Wang, J. & Dow, J. A. T. Using FlyAtlas to identify better Drosophila 760 melanogaster models of human disease. Nat. Genet. 39, 715–720 (2007). 761 63. Claes, L. et al. De novo mutations in the sodium-channel gene SCN1A cause severe 762 myoclonic epilepsy of infancy. Am. J. Hum. Genet. 68, 1327–1332 (2001). 763 64. Ardlie, K. G. et al. The Genotype-Tissue Expression (GTEx) pilot analysis: Multitissue 764 gene regulation in humans. Science 348, 648–660 (2015). 765 65. Ernst, C. Proliferation and Differentiation Deficits are a Major Convergence Point for 766 Neurodevelopmental Disorders. Trends Neurosci. 39, 290–299 (2016). 767 66. Marchetto, M. C. et al. Altered proliferation and networks in neural cells derived from 768 idiopathic autistic individuals. Mol. Psychiatry 22, 820–835 (2017). 769 67. Wei, H., Alberts, I. & Li, X. The apoptotic perspective of autism. Int. J. Dev. Neurosci. 770 36, 13–18 (2014). 771 68. Hartl, T. A. & Scott, M. P. Wing tips: The wing disc as a platform for studying Hedgehog 772 signaling. Methods 68, 199–206 (2014). 773 69. de Celis, J. F. & García-Bellido, A. Roles of the Notch gene in Drosophila wing 774 . Mech. Dev. 46, 109–122 (1994). 775 70. Raftery, L. A. & Umulis, D. M. Regulation of BMP activity and range in Drosophila wing 776 development. Curr. Opin. Cell Biol. 24, 158–65 (2012). 777 71. Diaz-Benjumea, F. J. & Cohen, S. M. Serrate signals through Notch to establish a 778 Wingless-dependent organizer at the dorsal/ventral compartment boundary of the 779 Drosophila wing. Development 121, 4215–4225 (1995). 780 72. Becam, I. & Milán, M. A permissive role of Notch in maintaining the DV affinity 781 boundary of the Drosophila wing. Dev. Biol. 322, 190–8 (2008). 782 73. Zecca, M., Basler, K. & Struhl, G. Sequential organizing activities of engrailed, hedgehog 783 and decapentaplegic in the Drosophila wing. Development 121, 2265–2278 (1995). 784 74. Tabata, T. & Kornberg, T. B. Hedgehog is a signaling protein with a key role in patterning 785 Drosophila imaginal discs. Cell 76, 89–102 (1994). 786 75. O’Roak, B. J. et al. Sporadic autism exomes reveal a highly interconnected protein 787 network of de novo mutations. Nature 485, 246–250 (2012). 788 76. Hahn, H. et al. Mutations of the human homolog of Drosophila patched in the nevoid 789 basal cell carcinoma syndrome. Cell 85, 841–851 (1996). 790 77. Johnson, R. L. et al. Human homolog of patched, a candidate gene for the basal cell nevus 791 syndrome. Science 272, 1668–1671 (1996). 792 78. Swarup, S., Pradhan-Sundd, T. & Verheyen, E. M. Genome-wide identification of 793 phospho-regulators of Wnt signaling in Drosophila. Development 142, 1502–1515 (2015). 794 79. Six, E. M. et al. The Notch Delta1 recruits Dlg1 at cell-cell contacts and regulates 795 cell migration. J. Biol. Chem. 279, 55818–55826 (2004). 796 80. Greene, C. S. et al. Understanding multicellular function and disease with human tissue- 797 specific networks. Nat. Genet. 47, 569–576 (2015).

33

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

798 81. Bult, C. J. et al. Mouse Genome Database (MGD) 2019. Nucleic Acids Res. 47, D801– 799 D806 (2019). 800 82. Zhou, W. et al. GluR1 controls dendrite growth through its binding partner, SAP97. J. 801 Neurosci. 28, 10220–10233 (2008). 802 83. Iizuka-Kogo, A., Ishidao, T., Akiyama, T. & Senda, T. Abnormal development of 803 urogenital organs in Dlgh1-deficient mice. Development 134, 1799–1807 (2007). 804 84. Mahoney, Z. X. et al. Discs-large homolog 1 regulates smooth muscle orientation in the 805 mouse ureter. Proc. Natl. Acad. Sci. 103, 19872–19877 (2006). 806 85. Caruana, G. & Bernstein, A. Craniofacial dysmorphogenesis including cleft palate in mice 807 with an insertional in the discs large gene. Mol. Cell. Biol. 21, 1475–83 (2001). 808 86. Chapman, D. L. & Papaioannou, V. E. Three neural tubes in mouse embryos with 809 mutations in the T-box gene Tbx6. Nature 391, 695–697 (1998). 810 87. Ashburner, M. et al. Gene ontology: Tool for the unification of biology. Nat. Genet. 25, 811 25–29 (2000). 812 88. The Gene Ontology Consortium. The Gene Ontology Resource: 20 years and still GOing 813 strong. Nucleic Acids Res. 47, D330-D338 (2019). 814 89. Pabis, M., Neufeld, N., Shav-Tal, Y. & Neugebauer, K. M. Binding properties and 815 dynamic localization of an alternative isoform of the cap-binding complex subunit CBP20. 816 Nucleus 1, 412–421 (2010). 817 90. Di Padova, M. et al. Che-1 arrests human colon carcinoma cell proliferation by displacing 818 HDAC1 from the p21WAF1/CIP1 promoter. J. Biol. Chem. 278, 36496–504 (2003). 819 91. Bruno, T. et al. Che-1 affects cell growth by interfering with the recruitment of HDAC1 820 by Rb. Cancer Cell 2, 387–399 (2002). 821 92. Page, G., Lödige, I., Kögel, D. & Scheidtmann, K. H. AATF, a novel transcription factor 822 that interacts with Dlk/ZIP kinase and interferes with apoptosis. FEBS Lett. 462, 187–91 823 (1999). 824 93. Sumiyoshi, E., Sugimoto, A. & Yamamoto, M. Protein phosphatase 4 is required for 825 centrosome maturation in mitosis and sperm meiosis in C. elegans. J. Cell Sci. 115, 1403– 826 1410 (2002). 827 94. Jensen, M. & Girirajan, S. An interaction-based model for neuropsychiatric features of 828 copy-number variants. PLoS Genet. 15, e1007879 (2019). 829 95. Coe, B. P., Girirajan, S. & Eichler, E. E. A genetic model for neurodevelopmental disease. 830 Curr. Opin. Neurobiol. 22, 829–836 (2012). 831 96. Nicholas, A. K. et al. The molecular landscape of ASPM mutations in primary 832 microcephaly. J. Med. Genet. 46, 249–253 (2009). 833 97. Brand, A. H. & Perrimon, N. Targeted as a means of altering cell fates 834 and generating dominant phenotypes. Development 118, 401–15 (1993). 835 98. Green, E. W., Fedele, G., Giorgini, F. & Kyriacou, C. P. A Drosophila RNAi collection is 836 subject to dominant phenotypic effects. Nat. Methods 11, 222–223 (2014). 837 99. Vissers, J. H. A., Manning, S. A., Kulkarni, A. & Harvey, K. F. A Drosophila RNAi 838 library modulates Hippo pathway-dependent tissue growth. Nat. Commun. 7, 10368 839 (2016). 840 100. Schindelin, J. et al. Fiji: An open-source platform for biological-image analysis. Nat. 841 Methods 9, 676–682 (2012). 842 101. Kanehisa, M., Sato, Y., Furumichi, M., Morishima, K. & Tanabe, M. New approach for 843 understanding genome variations in KEGG. Nucleic Acids Res. 47, D590–D595 (2019).

34

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

844 102. Hagberg, A. A., Schult, D. A. & Swart, P. J. Exploring network structure, dynamics, and 845 function using NetworkX. in 7th Python in Science Conference (SciPy 2008) 11–15 846 (2008). 847 103. Mi, H. et al. PANTHER version 11: Expanded annotation data from Gene Ontology and 848 Reactome pathways, and data analysis tool enhancements. Nucleic Acids Res. 45, D183– 849 D189 (2017). 850 104. Shannon, P. et al. Cytoscape: A software Environment for integrated models of 851 biomolecular interaction networks. Genome Res. 13, 2498–2504 (2003). 852 105. Köhler, S. et al. Expansion of the Human Phenotype Ontology (HPO) knowledge base and 853 resources. Nucleic Acids Res. 47, D1018–D1027 (2019). 854

35

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

855 FIGURE LEGENDS

856 Figure 1. Phenotypic expression of CNV carriers across tissues. Heatmap with frequencies of

857 non-neuronal developmental phenotypes observed in 1,225 human carriers of 10 pathogenic

858 CNV deletions, curated from the DECIPHER database, is shown. CNV carriers show a variety of

859 phenotypes that manifest across different tissues, including eye, limbs, muscle, and skeleton.

860

861 Figure 2. Targeted analysis to identify global developmental phenotypes with knockdown

862 of homologs of CNV genes. (A) Strategy for identifying non-neuronal phenotypes and

863 underlying cellular mechanisms for homologs of CNV and known neurodevelopmental genes

864 using the fly wing as a model system. We evaluated 59 Drosophila homologs of genes within 10

865 CNV regions and 20 known neurodevelopmental genes (79 total homologs). Using the UAS-

866 GAL4 system with wing-specific bxMS1096 driver, we knocked down 136 individual RNAi lines

867 for the CNV and neurodevelopmental homologs, and evaluated qualitative and quantitative

868 phenotypes. We next clustered RNAi lines based on severity of qualitative phenotypes, and

869 compared adult wing phenotypes to phenotypes observed with ubiquitous and eye-specific

870 knockdown of homologs. Furthermore, we evaluated underlying cellular mechanisms for the

871 observed wing-specific phenotypes, and examined the connectivity patterns of candidate

872 homologs for developmental phenotypes in multiple human tissue-specific networks. (B)

873 Representative brightfield images of adult wing phenotype severity observed with knockdown of

874 homologs of CNV genes, based on clustering analysis, are shown. (C) Heatmap with k-means

875 clustering of qualitative phenotypes in adult female wings across 136 RNAi lines is shown. The

876 color of each cell represents the frequency of individual fly wings (n=20-25 adult wings) for

877 each RNAi line (x-axis) that show a specific severity (no phenotype, mild, moderate, severe,

36

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

878 lethal) for the five qualitative phenotypes assessed (y-axis; wrinkled wings, ectopic veins,

879 missing veins, discoloration, bristle planar polarity), as detailed in Supp. Data 2. Based on these

880 data, we identified clusters for no phenotype (n=75 lines), mild (n=24 lines), moderate (n=10

881 lines), severe (n=21 lines), and lethal (n=6 lines). (D) Summary table of qualitative and

882 quantitative adult wing phenotypes for all tested RNAi lines of homologs of CNV and

883 neurodevelopmental genes. Quantitative phenotype totals do not include lethal RNAi lines for

884 both area and vein length. In addition, L3 vein length totals do not include severe RNAi lines.

885

886 Figure 3. Qualitative adult wing phenotypes of Drosophila homologs of CNV and

887 neurodevelopmental genes. Heatmaps representing the five qualitative adult wing phenotypes

888 for all 136 RNAi lines, with (A) all 59 tested homologs for 10 CNV regions and (B) 20

889 homologs for neurodevelopmental genes (β-catenin, core neurodevelopmental genes, and

890 microcephaly genes), are shown. The color of each cell represents the frequency of each of the

891 five qualitative phenotypes by severity (wrinkled wings, WR; ectopic veins, EV; missing veins,

892 MV; discoloration, DC; bristle planar polarity, BP), ranging from no phenotype to lethal. (C)

893 Representative brightfield images of adult fly wings (scale bar = 500µm) with wing-specific

894 knockdown of homologs of CNV and neurodevelopmental genes showing the five assessed

895 qualitative phenotypes, including discoloration, wrinkled wings, bristle polarity, ectopic veins,

896 and missing veins are shown. The panels in the bxMS1096-GAL4 control and C6836KK112485 images

897 highlight bristle planar polarity phenotypes for the representative images. Black arrowheads

898 highlight ectopic veins and white arrowheads highlight missing veins. Genotypes for the images

899 are: w1118/bxMS1096-GAL4;+; UAS-Dicer2/+, w1118/bxMS1096-GAL4;UAS-RphGD7330 RNAi/+;UAS-

900 Dicer2/+, w1118/bxMS1096-GAL4;UAS-CG15528KK107736 RNAi/+; UAS-Dicer2/+, w1118/bxMS1096-

37

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

901 GAL4;UAS-CG6836KK112485 RNAi/+; UAS-Dicer2/+, w1118/bxMS1096-GAL4;+;UAS-

902 CG14182GD2738 RNAi/UAS-Dicer2, and w1118/bxMS1096-GAL4;UAS-kisKK100890 RNAi/+; UAS-

903 Dicer2/+.

904

905 Figure 4. Quantitative adult wing phenotypes of Drosophila homologs of CNV and

906 neurodevelopmental genes. (A) Representative brightfield images of adult fly wings (scale bar

907 = 500µm) with wing-specific knockdown of homologs of CNV and neurodevelopmental genes

908 with size defects are shown. The bxMS1096-GAL4 control image highlights the six veins, including

909 longitudinal veins L2, L3, L4, and L5 as well as the anterior and posterior crossveins (ACV and

910 PCV), that were measured for quantitative analysis. The dotted line in the control image

911 represents the total wing area calculated for each RNAi line. Genotypes for the images are:

912 w1118/bxMS1096-GAL4;+; UAS-Dicer2/+, w1118/bxMS1096-GAL4;UAS-Fmo-2KK109203 RNAi/+; UAS-

913 Dicer2/+, and w1118/bxMS1096-GAL4;+;UAS-TrpmGD4541 RNAi/UAS-Dicer2. (B) Boxplot of L3

914 vein lengths for knockdown of select homologs in adult fly wings (n = 9-91, *p < 0.05, two-

915 tailed Mann–Whitney test with Benjamini-Hochberg correction). Vein measurements for all

916 other longitudinal veins and crossveins (ACV and PCV) for these lines are represented in Supp

917 Fig. 2. (C) Boxplot of wing areas for knockdown of select homologs in adult fly wings (n = 9-

918 91, *p < 0.05, two-tailed Mann–Whitney test with Benjamini-Hochberg correction). All boxplots

919 indicate median (center line), 25th and 75th percentiles (bounds of box), and minimum and

920 maximum (whiskers), with red dotted lines representing the control median.

921

922 Figure 5. Comparison of wing-specific, eye-specific, and ubiquitous knockdown of

923 homologs of CNV and known neurodevelopmental genes. (A) Heatmap with the penetrance

38

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

924 of phenotypes with ubiquitous knockdown (da–GAL4) of select homologs of CNV genes,

925 compared to their adult wing-specific (bxMS1096–GAL4) phenotypic severity is shown. (B)

926 Boxplots of Flynotyper-derived phenotypic scores for adult eyes with eye-specific knockdown

927 (GMR-GAL4) of select homologs of CNV and neurodevelopmental genes, normalized as fold-

928 change (FC) to control values (n = 7–40, *p < 0.05, one-tailed Mann–Whitney test with

929 Benjamini-Hochberg correction). The boxplots are arranged by severity of adult wing

930 phenotypes observed for each RNAi line, while the Flynotyper phenotypic scores are categorized

931 into four severity categories: no change (0–1.1 FC), mild (1.1–1.5 FC), moderate (1.5–2.0 FC),

932 and severe (>2.0 FC). (C) Boxplot showing the average eye phenotypic scores for 66 RNAi lines

933 of select homologs of CNV and neurodevelopmental genes, normalized as fold-change (FC) to

934 control values, by wing phenotypic category (n=4–30 RNAi lines per group). We did not observe

935 any significant changes in eye phenotype severity across the five wing phenotypic categories

936 (Kruskal-Wallis rank sum test, p=0.567, df = 5, χ2 = 3.881). Examples of average eye phenotypic

937 scores for RNAi lines with no phenotype (paraGD3392_1), mild (rlKK115768), and lethal (dlg1GD4689)

938 wing phenotype severity are highlighted in the graph. All boxplots indicate median (center line),

939 25th and 75th percentiles (bounds of box), and minimum and maximum (whiskers), with red

940 dotted lines representing the control median. (D) Representative brightfield adult eye (scale

941 bar = 100 µm) and adult wing (scale bar = 500µm) images with tissue-specific knockdown of

942 homologs of CNV genes are shown. Genotypes for the eye images are: w1118;GMR-GAL4/+;

943 UAS-Dicer2/+, w1118;GMR-GAL4/UAS-LnkKK105731 RNAi; UAS-Dicer2/+, w1118;GMR-

944 GAL4/UAS-mEFTu1GD16961 RNAi; UAS-Dicer2/+. Genotypes for the wing images are:

945 w1118/bxMS1096-GAL4;+; UAS-Dicer2/+, w1118/bxMS1096-GAL4; UAS-LnkKK105731 RNAi/+; UAS-

946 Dicer2/+, and w1118/bxMS1096-GAL4; UAS-mEFTu1GD16961 RNAi/+; UAS-Dicer2/+.

39

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

947 Figure 6. Expression patterns of Drosophila homologs and human CNV and

948 neurodevelopmental genes across multiple tissues. (A) Heatmap with expression of fly

949 homologs of select CNV and neurodevelopmental genes in multiple Drosophila larval and adult

950 tissues, derived from the FlyAtlas Anatomical Microarray dataset, compared with adult wing

951 phenotype severity, is shown. Expression values are grouped into no expression (<10 fragments

952 per kilobase of transcript per million reads, or FPKM), low (10–100 FPKM), moderate (100–500

953 FPKM), high (500–1000 FPKM), and very high (>1000 FPKM) expression categories. (B)

954 Heatmap with expression of select human CNV and neurodevelopmental genes in multiple adult

955 tissues, derived from the Genotype-Tissue Expression (GTEx) dataset v.1.2, is shown.

956 Expression values are grouped into no expression (<3 transcripts per million reads, or TPM), low

957 (3–10 TPM), moderate (10–25 TPM), high (25–100 TPM), and very high (>100 TPM)

958 expression categories. X symbols denote preferential expression in a particular tissue (see

959 Methods). Expression data for all CNV and neurodevelopmental genes are provided in Supp.

960 Data 6.

961

962 Figure 7. Drosophila homologs of CNV and neurodevelopmental genes show altered levels

963 of apoptosis and proliferation. (A) Larval imaginal wing discs (scale bar = 50 µm) stained with

964 nuclear marker DAPI, apoptosis marker dcp1, and cell proliferation marker pH3 illustrate altered

965 levels of apoptosis and cell proliferation due to wing-specific knockdown of select fly homologs

966 of CNV genes. We quantified the number of stained cells within the wing pouch of the wing disc

967 (white box), which becomes the adult wing. Additional representative images of select homologs

968 are presented in Supp Fig. 5. Genotypes for the wing images are: w1118/bxMS1096-GAL4;+; UAS-

969 Dicer2/+, w1118/bxMS1096-GAL4;+; UAS-AatfGD7229 RNAi/UAS-Dicer2, w1118/bxMS1096-GAL4;UAS-

40

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

970 Pp4-19CGD9561/+; UAS-Dicer2/+, w1118/bxMS1096-GAL4;+; UAS-Atx2GD11562 RNAi/UAS-Dicer2,

971 and w1118/bxMS1096-GAL4;+; UAS-SinGD7027 RNAi/UAS-Dicer2. (B) Box plot of dcp1-positive

972 cells in larval wing discs with knockdown of select fly homologs of CNV and

973 neurodevelopmental genes, normalized to controls (n = 7–18, *p < 0.05, two-tailed Mann–

974 Whitney test with Benjamini-Hochberg correction). We note that several RNAi lines showed

975 severe dcp1 staining across the entire wing disc and could not be quantified. The number of dcp1

976 positive cells were calculated manually. (C) Box plot of pH3-positive cells in the larval wing

977 discs with knockdown of select fly homologs of CNV and neurodevelopmental genes,

978 normalized to controls (n = 6–18, *p < 0.05, two-tailed Mann–Whitney test with Benjamini-

979 Hochberg correction). The number of pH3 positive cells were calculated using the

980 AnalyzeParticles function in ImageJ. All boxplots indicate median (center line), 25th and 75th

981 percentiles (bounds of box), and minimum and maximum (whiskers), with red dotted lines

982 representing the control median.

983

984 Figure 8. Candidate Drosophila homologs of genes within CNV regions interact with

985 conserved signaling pathways. Larval imaginal wing discs (scale bar = 50 µm) stained with (A)

986 wingless, (B) patched, (C) engrailed, and (D) delta illustrate disrupted expression patterns for

987 proteins located within the Wnt (wingless), Hedgehog (patched and engrailed), and Notch (delta)

988 signaling pathways due to wing-specific knockdown of select fly homologs of CNV and

989 neurodevelopmental genes. Dotted yellow boxes represent expected expression patterns for

990 signaling proteins in bxMS1096-GAL4 control images. White arrowheads and dotted white boxes

991 highlight disruptions in expression patterns of signaling proteins with knockdown of CNV or

992 neurodevelopmental genes. Additional representative images of select homologs are presented in

41

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

993 Supp Fig. 7. Genotypes for the wing images are: w1118/bxMS1096-GAL4;+; UAS-Dicer2/+,

994 w1118/bxMS1096-GAL4;UAS-Pp4-19CGD9561/+; UAS-Dicer2/+, w1118/bxMS1096-GAL4;UAS-

995 Cbp20KK109448/+; UAS-Dicer2/+, w1118/bxMS1096-GAL4;+; UAS-SinGD7027 RNAi/UAS-Dicer2,

996 w1118/bxMS1096-GAL4;+; UAS-AatfGD7229 RNAi/UAS-Dicer2, and w1118/bxMS1096-GAL4;UAS-

997 Klp61FGD14149/+; UAS-Dicer2/+.

998

999 Figure 9. Connectivity of human CNV genes with conserved signaling pathway genes in

1000 human tissue-specific networks. (A) Representative diagrams of eight human CNV and

1001 neurodevelopmental genes whose fly homologs disrupt the and 57

1002 human Notch signaling genes within kidney, heart, and brain-specific gene interaction networks

1003 are shown. Yellow nodes represent CNV and neurodevelopmental genes, pink nodes represent

1004 Notch signaling pathway genes, and green nodes represent connector genes within the shortest

1005 paths between CNV and Notch pathway genes. (B) Violin plots showing the average

1006 connectivity (i.e. inverse of shortest path lengths) of CNV genes to genes in Hedgehog, Notch,

1007 and Wnt signaling pathways across the tested tissue-specific networks (n=322–810 pairwise

1008 interactions, *p < 0.05, two-tailed Welch’s t-test with Benjamini-Hochberg correction). (C)

1009 Table showing enriched clusters of Gene Ontology (GO) Biological Process terms for connector

1010 genes observed for each signaling pathway in the three tested tissue-specific networks,

1011 categorized by enrichments in ubiquitous, neuronal, and non-neuronal tissues (p<0.05, Fisher’s

1012 Exact test with Benjamini-Hochberg correction).

1013

1014 Figure 10. A multigenic model for neuronal and non-neuronal phenotypes associated with

1015 pathogenic CNVs. Schematic of a multigenic model for neuronal and non-neuronal phenotypes

42

bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

1016 associated with pathogenic CNVs. While a subset of genes within CNV regions contribute

1017 towards tissue-specific phenotypes, a majority of genes contribute towards both neuronal and

1018 non-neuronal phenotypes through disruption of developmental signaling pathways and global

1019 biological processes.

43

Distal 16p11.2deletion bioRxiv preprint not certifiedbypeerreview)istheauthor/funder,whohasgrantedbioRxivalicensetodisplaypreprintinperpetuity.Itmadeavailable 16p13.11 deletion 16p11.2 deletion 16p12.1 deletion 15q13.3 deletion 15q11.2 deletion 7q11.23 deletion All CNVCarriers Figure 1 1q21.1 deletion 17q12 deletion doi: 3q29 deletion https://doi.org/10.1101/855338 (n= 1225) (n= 251) (n= 192) (n= 249) (n= 118) (n= 61) (n= 63) (n= 79) (n= 80) (n= 92) (n= 40) under a ; this versionpostedNovember26,2019.

Cardiovascular CC-BY 4.0Internationallicense Craniofacial Digestive system Ear . The copyrightholderforthispreprint(whichwas Eye Genitourinary Growth Integument Limbs Muscle Skeleton frequency Phenotype 0.0 0.1 0.2 0.3 0.4 0.5 bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

Figure 2

A Experimental strategy B Phenotypic analysis of adult wings in Drosophila × bxMS1096-GAL4 control No phenotype Mild Moderate Severe Wing-specific driver UAS-RNAi bxMS1096-GAL4 (79 genes)

C K-means clustering of qualitative wing data Phenotyping of adult wings Qualitative analysis Quantitative analysis No observable phenotype Mild Moderate Severe Lethal Frequency (n=75) (n=24) (n=10) (n=21) (n=6) • Wrinkled wings • Longitudinal veins 1.0 • Ectopic veins (L2,L3,L4,L5) 0.8 • Missing veins • Crossveins (ACV, PCV) • Discoloration • Wing area 0.6 Bristle planar polarity • 0.4

0.2 Tissue-specific effects Phenotypic severity 0.0 • Compare to ubiquitous knockdown 136 Drosophila RNAi lines • Compare to eye-specific knockdown • Gene expression across multiple tissues (Human and flies) D Summary of qualitative and quantitative adult wing phenotypes

Cellular processes and Qualitative phenotypes Quantitative phenotypes Fly homologs RNAi lines developmental pathways No pheno. Mild Moderate Severe Lethal Wing area L3 vein length • Apoptosis CNV genes • Cell proliferation (15q11.2, 15q13.3, 16p11.2, Distal 16p11.2, 16p12.1, 16p13.11, 17q12, 1q21.1, 3q29, 7q11.23) 95 45 21 7 16 6 65 53 • Disruptions in signaling pathways Known neurodevelopmental genes • Human tissue-specific network analysis (β-catenin, core genes, microcephaly) 41 30 3 3 5 0 24 21 (Brain, heart, kidney) Total 136 7524 10 21 6 89 74 bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

Figure 3 A Qualitative wing phenotypes of homologs of CNV genes 15q11.2 15q13.3 16p11.2 Distal 16p11.2 16p12.1 16p13.3 17q12 1q21.1 3q29 7q11.23 BP MV DC WR Lethal EV BP MV DC WR Severe EV BP MV DC

Mod. WR EV BP MV DC Mild WR EV BP MV DC WR EV No pheno. GD4689 GD7229 GD3061 GD9561 GD6674 GD9689 GD2249 GD1241 GD6991 GD6343 GD3482 GD9362 GD4372 GD2442 GD9322 GD9260 GD7330 GD6560 GD9986 GD7027 GD15691 GD16961 GD11699 GD564_1 GD11803 GD11383 GD11562 GD564_2 GD15226 GD17096 GD13375 GD14217 GD10067 KK104454 KK105731 KK102281 KK108714 KK100134 KK108281 KK101874 KK101201 KK108049 KK107404 KK107824 KK102573 KK108623 KK100453 KK104813 KK109203 KK102092 KK108812 KK108732 KK104762 KK115768 KK109448 KK107680 KK102370 KK108740 KK101936 KK105756 KK109046 KK108466 KK107736 KK116285 KK107305 KK112485 KK110317 KK109207 KK107577 KK108227 KK109717 KK108387 KK101457 GD8318_1 GD4541_1 GD1138_1 GD2738_1 GD3777_1 GD9586_1 GD1266_1 GD2856_1 GD8318_2 GD2738_2 GD4541_2 GD1266_2 GD2856_2 GD1138_2 GD3777_2 GD9586_2 GD11477_1 GD14152_1 GD11238_1 GD14152_2 GD11477_2 GD11238_2 ke r l lgs Sin ke Lnk Pa1 Aatf lgs Cen Rph dlg1 Tsf2 Pis Fsn Sin ACC Lnk shu Lim1 app Pak app Trpm coro Pa1 Ald1 Atx2 Aatf spin dlg1 Pcyt1 Ald1 Marf1 spict nudE a−1 a−1 Lim1 MRP nudE Doc2 Nsun5 Trpm Trpm Sgf29 Pcyt1 Marf1 Syx1A Syx1A Sgf29 Nsun5 Ada2a Nsun5 PIG−Z Cbp20 LIMK1 LIMK1 PIG−X Sr Sr Fmo−2 CG9150 Fmo−2 Fmo−2 PIG−Wb mEFTu1 CG5589 Pp4−19C mEFTu1 CG2059 CG15528 CG5589 CG8204 CG6836 CG8888 CG8888 Grip128 Grip128 BCL7−li α 5 nAChR CG15309 CG11035 CG17841 CG10465 CG15528 CG14411 CG14182 CG11035 CG14411 CG11035 CG14182 α 5 nAChR α 5 nAChR CLIP−190 BCL7−li UQCR−C2 UQCR−C2 UQCR−C2 B Qualitative wing phenotypes of homologs C Representative qualitative phenotypes of neurodevelopmental genes in the adult wing Frequency β-catenin genes Core genes Microcephaly bxMS1096-GAL4 RphGD7330 BP 1.0 MV DC WR Lethal EV BP 0.8 MV DC

WR Discoloration (DC) Severe EV 0.6 BP KK112485 CG14182GD2738_1 MV CG6836 DC

Mod. WR EV 0.4 BP MV DC Mild WR EV

0.2 Ectopic vein (EV) BP Bristle polarity (BP) MV DC CG15528KK107736 kisKK100890 WR EV

No pheno. 0.0 GD39 GD15 GD2619 GD9974 GD1372 GD6852 GD2905 GD2535 rk GD450_1 GD14481 GD11734 GD10101 GD450_2 GD13500 GD12537 KK108009 KK108534 KK109063 KK101542 KK100878 KK109278 KK100890 KK102545 KK107895 KK101537 KK108197 GD3392_1 GD1375_1 GD6121_1 GD9502_1 GD3392_3 GD3392_2 GD6121_2 GD9502_2 GD1375_2 GD14451_1 GD14383_1 GD14149_1 GD14383_2 GD14451_2 GD14149_2 Eph rk arm kis Eph rk rk Wrinkled (WR) Wrinkled Eph Gat Tpc1 asp asp arm Pten para Pten para para para Nrx−1 Sas−4 Missing vein (MV) org−1 Ube3a Ube3a Sas−4 Prosap MCPH1 Cadps Cadps Scs β G Nrx−1 Nrx−1 Prosap MCPH1 Grip163 Grip163 Klp61F Klp61F Cep135 Cep135 bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

Figure 4 A Representative quantitative phenotypes in the adult wing Smaller area/L3 vein length Larger area/L3 vein length L5

PCV L4 ACV L3

L2 bxMS1096-GAL4 Fmo-2KK109203 TrpmGD4541_2

B L3 vein lengths for select homologs C Wing area for select homologs epha ly epha ly gen es -catenin -catenin q21 .1 5q13 .3 q21 .1 5q13 .3 q2 9 q11 .23 6p11 .2 q2 9 q11 .23 6p11 .2 6p1 3.11 5q11 .2 5q11 .2 1 3 7 1 1 17q12 β Core Genes Microc 1 3 7 1 1 1 β Core Microc 1 1 17q12 Distal 16p11.2 Distal 16p11.2 2500 20 * * ** * * * * * * *

) 15 * * 2 * * * * * * 2000 * * * * * * * * * µm * 5 * * * * * * * 10 * *

Le ngth ( μ m) *

1500 Area (×10 * 5 * * * *

1000 0 124 1 656 0 124 1 656 0 AL 4 AL 4 1156 2 1138 3 1709 6 1006 7 1375 _1 3777 _1 9586 _1 4541 _2 4541 _2 1375 _1 GD GD GD GD -G 14149 _2 11477 _2 -G 11477 _2 14451 _2 GD GD GD GD KK 1092 03 KK 1094 48 KK 1018 74 KK 1087 32 KK 1057 31 KK 1157 68 KK 1087 14 KK 1014 57 KK 1004 53 KK 1092 78 KK 1015 37 KK 1081 97 KK 1008 90 KK 1092 03 KK 1124 85 KK 1018 74 KK 1087 32 KK 1057 31 KK 1025 73 KK 1157 68 KK 1087 14 KK 1084 66 KK 1092 78 KK 1015 37 KK 1081 97 KK 1008 90 GD GD GD GD GD GD GD GD lgs GD GD lgs rl rl kis kis Fsn 91 50 91 50 Atx2 Pa k asp Ln k Pa k asp Ln k Aa tf Pten Pten 109 6 MS 20 59 bp 20 109 6 MS 68 36 20 59 82 04 org-1 88 MRP Sg f29 org-1 Trpm Trpm un 5 Ns 153 09 178 41 153 09 Fmo-2 p6 1F Fmo-2 C Sra-1 LIMK1 CG Nrx-1 Sra-1 CG Prosap Prosap bx bx CG CG CG CG Kl CG CG CG CG bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

Figure 5

A Ubiquitous knockdown B Eye versus wing phenotypes

No wing Wing measurement Mild wing Mod. wing Sev. wing phenotype phenotype phenotype phenotype phenotype Lethal 3 * * * * * * Severe * * * * * Fly RNAi line LarvalPupal lethalityWing lethality Nodefects gross defectsWing phenotypeCNV region sco re (FC) 2 * * * Moderate Sra−1GD11477_2 ns

KK102281 15q11.2 yp ic ns spict Mild Grip128GD14152_1 1 CG14411GD8318_2 GD9986 15q13.3 Trpm phen ot No change Ald1KK107680 KK109207 Pis Eye 0 coroGD14217 GD7330 290 5 261 9 124 1 273 8 733 0 468 9

Rph 1350 0 1696 1 KK102573 GD GD GD GD GD GD

CG17841 GD GD KK107577 KK105756 16p11.2 KK 1048 13 KK 1057 56 KK 1057 31 KK 102573 KK 1157 68 KK 1094 48 KK 1088 12 rl CG10465 lgs Rph KK115768 dlg1 Tpc1 Nrx-1 GMR-GAL4 Lnk Pten

rl spin 141 82

GD9561 bp 20 Sgf29

Pp4−19C 104 65 C KK108623 CR -C2 mEFTu1 Doc2 CG CG17841 KK108714 CG CG15309 UQ Atx2GD11562 Sgf29KK107577 C Severity of eye D Representative eye and wing images LnkKK105731 Distal spinKK104813 16p11.2 versus wing phenotype GMR-GAL4 bxMS1096-GAL4 mEFTu1GD16961 3 lgsGD1241 ns KK109203 1q21.1 Fmo−2 KK115768 CG8888GD3777_1 rl dlg1GD4689 FsnGD11383 2 dlg1GD4689 Control Tsf2GD2442 Cbp20KK109448 3q29 sc ore (FC) CG6836KK112485 PakKK101874

KK109717 Average e ye

PIG−X no typic 1 KK107404 PIG−Z GD3392_1

para KK105731 Ubiquitous Wing ph e Lnk Ubiquitous (da-GAL4) Wing-specific MS1096 0 Full penetrance (bx -GAL4)

Partial penetance Lethal Mild

No phenotype Le thal GD16961 Severe Se vere ode rate

Moderate he notype M Mild phenotype

Wing size changes Measurement No p

No phenotype mEFTu1 Wing phenotypic severity Figure 6 Larval Adult CNV regions bioRxiv preprint not certifiedbypeerreview)istheauthor/funder,whohasgrantedbioRxivalicensetodisplaypreprintinperpetuity.Itmadeavailable Drosophila Drosophila (FlyAtlas) (FlyAtlas) Muscle Kidney Heart Brain Lung Liver Wing phenotype B A doi:

CYFIP1 15q11.2 https://doi.org/10.1101/855338 Fat body Fat body Expression patternsofflyhomologs ofCNVgenesacrossmultipleflytissues

NIPA2 Trachea CHRNA7

15q13.3 Heart Brain

X X TRPM1 CNS Gut Gut

X ALD O A

X CORO1A DOC2A FAM57B Expression patternsofCNVgenesacrossmultiplehumantissues KCTD13 16p11.2 MRP MAPK3 Cen

X X PPP4C spin

QPRT under a asp ; this versionpostedNovember26,2019. X YPEL3 Cadps ATP2A1 Sgf29 ATXN2L CC-BY 4.0Internationallicense SH2B1 Distal 16p11.2 CG9150 SPNS1 Fsn TUFM CG10465 CCDC101 Lim1 CDR2 Nrx−1 16p12.1 PIG−X

X X POLR3E Pten

X X X X UQCRC2 ABCC6 para MYH11 16p13.11 Lnk DHRS11 Trpm AATF Ube3a DDX52 17q12 Fmo−2 . LHX1 CG2059 The copyrightholderforthispreprint(whichwas

X X ACACA lgs HNF1B CG8888 BCL9 1q21.1 CLIP−190 X X X FMO5 CG11035

X GJA5 BDH1 CG17841 DLG1 LIMK1 FBXO45 rl MFI2 org−1 NCBP2 3q29 CG15309 PAK2 BCL7−like Sra−1 X PIGX kis

X TFRC ABHD11 Rph BCL7B Pak CLIP2 Aatf DNAJC30 7q11.23 X Atx2 ELN nAChRα5

X LIMK1 arm

X X STX1A CG5589 CLDN3 Klp61F GTF2IRD1 Tsf2 CTNNB1 β-catenin Cbp20 NRXN1 Pp4−19C

X PTEN TBX1 mEFTu1 CADPS2 UQCR−C2 Core genes ACC

X CHD8 Sin

X SCN1A UBE3A dlg1

ASPM Microcephaly Expression Wing phenotype KIF11 Mild No phenotype No expression Low Moderate High Very high Wing sizechanges Moderate Severe Lethal Expression No expression Low Moderate High Very high A Figure 7 SinGD7027 Atx2GD11562 Pp4-19CGD9561 AatfGD7229 bxMS1096-GAL4 DAPI Cellular processesinlarvalwingdiscs bioRxiv preprint not certifiedbypeerreview)istheauthor/funder,whohasgrantedbioRxivalicensetodisplaypreprintinperpetuity.Itmadeavailable doi: https://doi.org/10.1101/855338 dcp1 ppoi Cellproliferation Apoptosis Increased Increased Increased Increased pH3 under a ; this versionpostedNovember26,2019. CC-BY 4.0Internationallicense Decreased Decreased Decreased Decreased

Normalized pH3 Normalized dcp1 C B positive cells positive cells 0.0 0.5 1.0 1.5 0 1 2 3 4 5 .

MS1096 The copyrightholderforthispreprint(whichwas bx -GAL4 (GD) bxMS1096-GAL4 (GD) MS1096 bx -GAL4 (KK) MS1096 * bx -GAL4 (KK) armKK102545 * KK109448 KK115768 * Cbp20 rl GD11562 * Atx2 KK105756 ns GD9561 * CG10465

Pp4-19C ns *

Quantification ofproliferatingcells GD3482 GD1241 ACC

lgs Quantification ofapoptoticcells * GD7229 GD11383 ns Aatf Fsn KK108197 * org-1 GD4689 ns LnkKK105731 * dlg1 * GD7330 KK101201 * Rph CG11035 KK107577 * Sgf29 KK107577 *

* Sgf29 Sra-1GD11477_2 *

* GD1241 rkKK108009 lgs * Klp61FGD14149_2 KK108009 * GD7027 * rk

Sin GD11477_2 * rl KK115768 * Sra-1 * CG11035KK101201 KK100890 *

ns kis KK109203 Fmo-2 GD11238_1 * ns UQCR-C2 UQCR-C2GD11238_1 ns KK108197 * CG15309 KK108714 org-1 KK102573 ns CG17841 KK108714 * sns ns CG15309 CG10465 KK105756 *

ns KK102573 ACCGD3482 CG17841

GD15226 KK109203 * nudE Fmo-2 KK100890 ns

kis KK105731 * ns Lnk FsnGD11383 * * GD3777_1 CG8888GD3777_1 CG8888 dlg1GD4689 * bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

Figure 8 Disruption of signaling pathways in larval wing discs bxMS1096-GAL4 Pp4-19CGD9561 Cbp20KK109448 SinGD7027 AatfGD7229 Klp61FGD14149_2 A Wnt wingless

B patched

C Hedgehog engrailed

D delta Notch bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

Figure 9 A Human CNV genes interact with Notch signaling pathway genes in multiple tissues Kidney Heart Brain

CTNNB1

DTX4

TCF3

CTNNA1 NDE1

GSK3B

SOCS5 ARF1

RBM39 MTA1

PHLDA1

MAML2

NR3C1

ATXN1L PAFAH1B1

IRF9

NUMBL

TIMP3

FOXO1

NDRG1 NCK1 ABL1

WBP11 PML NUMBL

SMAD3 KAT2A OS9 NOTCH2NL

CD44 DTX3L

HNRNPU

BZW1 CREB1

PSME3

TNS1

HES1 VIM

PSEN1

CANX TRIP6

ATXN1L

NRIP1

COL4A2 KAT2B

SIRT1

INPP5D

CIR1 PLSCR1

TMX1 MCL1

COPS5

TRIM27

TAB2 PTEN LYN

CDKN1B PDCD4

UBN1 HMGN4

TRIP10

PRR11

APP

EZR UBE2K MR1

HEY1 DVL1 FEZ1 SLK

TGIF1

STAT2

DLL4

HSPA4

DLL1 PSMC2

ATP2B4

NCOA3

KARS

RANBP2

CSNK1E PSMA1

RELA HLTF

CTDSP2 CREBBP

FMR1 EP300

NAB1 PPP2CA

PSMC6

CASK TM4SF1 HSP90AA1

LUC7L3

SMAD4

SMARCC1 CDC27

PRKD2

H2AFX

CEP170 PSMA1

CAMSAP2 FYN TLE2 KCTD12

CTCF LAPTM4B AZIN1

NARS NRIP1 UBR2 DAG1

PUF60

KAT6A UBE2D3 BUB3

NOTCH3

ATP2B4

TMED10

GFPT1 MORC3

NCOA3 VIM

LEPROTL1

SLC39A6

BHLHE40

FLNA NFKBIA NCOR1 RB1 NPR1 MAML2 NUMB CTBP1 ZNF146 PTDSS1 GRN PSEN2

MYO1B DDX1 DNAJB6

YY1

CSE1L COPS3

PTP4A2

BRCA1 TP53

QKI RBM10 SFSWAP PSENEN C1D

RAN

MKI67

DLL1 UTP6 P4HB PPP1R2

E2F1 DNAJA1 SRSF2

FNDC3B RAD54B LSM14A

BAZ2B HEYL SOCS5 CIR1 DPYSL2 CDK1 RANBP1

TNPO1 RBPJ

NOTCH2 NCSTN

DEK CTBP2 XPO1

PRPF3 CENPA PAXIP1

KAT2B ATXN1 MAML1 NOTCH1 SYNCRIP SRSF10

CCT5

DVL3 NCK1 NCOR2 DAZAP2 MFNG

CTBP2 SSRP1 SIRT1 PSMD14

CCT6A NAB1 SNW1

CIB1

JAG1

PA2G4

NOTCH2

APH1B DDX23 UBE2D2 ABCE1 FLNA SSR1 TRIOBP PUM2 CBX3

PAPOLA SSBP2 AKAP13 ATXN1 SET

GFPT1 PSMD14

DTX2 HDAC1

TUBB4B DVL2

RAB32 RBPMS

USP1 RASA1 PIK3CA ZC3H11A

PPP4R1

HFE RBBP8

SNW1

TLE4 CSNK1D

HES1 RHOXF2 RNF11

SIRT1 COPS2 KHSRP DKC1 PPP6C

BHLHE40

KIAA0355

NCBP2 NUP153 OAT ATF2 TPM4

NUP98 ABI1 IER2 RAB21 DDX39A NUP98 DTX2 AKT3 UBR5

PRPF4 DHX15

KHDRBS1 COPS3

NOTCH3

SNRPG QKI BCL6 HDAC2

FLAD1 ATP2B4

NARS ADAM10 TCF7L2

TLE1 HSPA4 NUMB HACD2 TLE4

ADAM17

RHOXF2 CDC20 PFN1 TGFB2 P4HA1 PARP1 DHX15 ITGB4

NOTCH1 MAD2L1 ARHGAP5

LAMP1 SRSF1 CCT5 DLG1 ILF3 SNRPD3

XRCC5 POLE2 ATP6V0B USP15 UBE2G1 NCK1

MTF2 KMT2A

PRPF40A MCM7 YES1 ARL1 COIL

RBPJ

PAXIP1 IL6ST

TLE4 UBR5

SNRPD1 EGFR SLC19A1

SAP30 SYT7

MAML2 TAGLN2 CYBRD1

CCT5 HDAC2

KDM2A ABL1 USP1

MAPRE1 SRSF3 HEY1

DAZAP2

XRCC6

PIK3R1 KDM5B

SERBP1 PPP1CA ADAM17 CDKN1A SMARCA4 EIF3A

RBPJ PRKDC PAPOLA CDC25A STK24 CTBP2

MAML1 RBPMS VEGFA EP300

IL6ST

MYC TLE1 HNRNPUL1 COIL DDR1 RAC1 MRPL3 PTBP1

LAMP1 TLE3 PSMD13 SMARCC1 CBFB CAVIN1

GSK3B DLG1 PJA2 NOTCH2 PTBP1

CCNA2 POLR3E

APH1A CDKN1A DEK

RB1CC1

CTNNA1 CLIC1 PPP4R1

JAG2 AATF NDE1 RSBN1

PAICS TOPBP1

CREBBP CALD1 WDR77

PUM2 SRSF2 BCAP31 EWSR1

GNA11 SNW1

EWSR1

LEPROTL1 TACC1 PTPN11 TUBG1 SMARCA5 NUDC

TLE2

NRP1

CKAP5

CREBBP PPP2R5C XRCC5 EP300

MRPL3 MAD2L1 SNRPG HDAC1 NRP2 TRIO GNB2 MAML1 NCBP2 NCOA3 SCARB2

AZIN1

RELA FYN ACTN1 JAG2 AATF PTBP1

HSPE1

SEC24A DKC1 DHX15

PDCD11 ACTL6A CTNNA1 HMOX2 YY1 TOPBP1 ATXN1

DDX1 TK1 CDC20 DNAJB6 USP1 XRCC6 NCBP2

DDR1 VARS UBR5

NUMB KIF11 ID4

JAG1 DLL1 UBE2N CCNB1

CIR1 CTBP1 RANBP9

TMED10 HDAC2

DNAJB6 CUL4B PRKDC RNPS1

ATP6V0D1

SERINC3 CSE1L TMEM109

UPF1

PA2G4 GMEB2

MCM5 TP53

JAG1 HSPA13 IL6ST GNA11

APLP2

PARP1 CCT7 SRSF2 CTBP1 TRIM2

ADAM17 CENPA PSEN1 SSRP1 DLG1

NCBP1 SEPT10 TPD52L2

BUB3 UBE2D4 TRIP13 CCT3 RELA ERBB2

ZMYM4 NOTCH1 DTX1 TRIP13 CCT3

SMAD4 PA2G4 SRP19 PDGFRA EIF3B STK24 ILF3

KAT2B BCS1L CCT7

ATIC TRA2B FLNA YWHAZ TPP1 PRPF3

GTSE1

YWHAZ

RUNX1 STAT3

MTF2 RAC1

HRAS HEY1 NUP62 MTDH PNN

EIF3A CCT3

KHDRBS1 PPP4C HEYL HSPA13 AGPAT2 RANBP9 CCT2 SUGP2 SYNCRIP GNB1 TTC3

TMEM97

TOPBP1

GMPS DKC1 NCSTN MAPRE1 ATP1B1

MORC3 PSME3 NCOR1

NUP85 POLR3E CDKN2C NCOR2 AZIN1 KDM5B

PPP1CA EIF2S1 DLL4 TIMP3

ATIC

EIF4G1

CREB1

UBE2D3

HDAC1 KDELR3

KDELR1 USP34

TPP1 DEK POLR3E HSP90AA1 KLC1 ATXN1L MAD2L1 TLE1

DAZAP2

SMARCA4 XRCC6 RFC3 KIF11 AIMP2

SELENOT

PRIM1

DIMT1 HNRNPA3 UBE2M GNB2 NDE1

CDK4 TROAP PARP1 EIF3B UBE2N HGS

CCNB1

UBE2D2 RNASEH2A C15orf39 DVL2 RNF11

GFPT1

CCNF

DLL3

PPP4C TRIM2 RFNG

TRIP13 CASK CDKN2C VARS CEP126 MSH6

CCT6A NUMBL PAICS

PSENEN

CANX

ACOT7 HRAS

SMARCA4 FUBP1 TMED10 SAP30

UBE2K DTX3 DVL3 HES1 CDK4 SRSF1 DDR1 RAD21 ILF3 SART3 TOMM40

CSE1L KAT2A

UBQLN1

APH1B

PKP4

TLE3 HRAS

BRCA1 RBBP7

AATF RNF126 DTX4

WDR77 PRPF3 UBQLN1 CCT7

ADRM1 HEY2

KAT2A CD151

BRCA1 RB1 PSEN2

UBE2D3 RNF138 NOTCH3 CTDSP2

CSNK1E HAP1

FNDC3B

ARFGAP1

HNRNPUL1

RNPS1 SSRP1 CSNK1D

POLE2

ADAM10

AGPAT2

GNB2 PPP4C RIN1 MBNL2 ILF2 TRIM27 UBE2D2

RUVBL2 NSD2 ILF2 CDC20 UPF3A TRIP6

VARS ANK3

CASP2 FKBP4 TCF3 PSEN2 BRD4

ESR1

PTPRK

GNA11 TMPO RAC1 RB1 BAG6 SFRP1 DVL1 KIF11 TLE3

DVL3 APH1B APH1A

DMWD CDK13 PPP2R1A SMAD3 HGS

UBE2H SPEN

PPP1CC CIB1

RANBP9 HSPA4 ARHGDIA

JAG2 MTUS1

NCOR2 TMBIM6 CSNK1G2 TAL1 PSEN1 PPP1CA SNRPA

TOP2A

DMWD MCM7 CXXC1

BOP1 NCSTN TRA2B UBE2K

TUBG1 BRF1 NONO RNPS1 GNB1 UBE2D4 RNF11 MCM5 NUDC

RFNG YWHAG

CUL4B JPT1 ALCAM APH1A

UBE2M

PBK

UBE2D4 CCNA2

HSPG2 UBE2M

CIB1

NPTXR

DTX3L

STRN4 RBPMS

ERBB4

CDK1 PSMB3

MTA1

EPHB4 DTX3

DTX1 TLE2

ATP6V0B

ADGRL1 SRP19 CTSC PSMB3

LTBP2

PTCRA BOP1

PPP2R1A TPX2 TOMM20

CCNA2

EWSR1 GRK2 PPP4R1

PSENEN MTA1 AKT2

UBE2H

PTCRA ZDHHC17

RECQL5 ATP6V0C LAMP1 MCM4

HGS MFI2 SYNCRIP CASP8

JAK3 ATIC DRD2

TRIM27

USP19

CCNF RARB ARL4D PLSCR1

RNF126

ARPC1A

DTX3L

MYCBP

ATP6V0C NFIB

TAOK2 GSK3B

RAC2 GTSE1 FEN1

MYO9B PHB

SNTA1

TP53

ARHGDIA RAD50

DTX3 ASXL1

MLF2

ATP6V0D1 MKI67

DTX4 CSNK1E

OTUB1

NOTCH4

VBP1

RCN2 TMBIM6 E2F1

EMP3

PAX8

CYP2A6

FTH1 ATN1

NCKIPSD

RFNG CASP10 DTX1

PLK1 TOMM40 DVL2 PSME2

RUNX2

EPS8L1

DDX11

BTRC CDC25B UBC

DVL1

FGF18

CDKN3 RAD54B BAG6

ARHGEF1

FZR1

MCRS1

NTRK3

GSTM5 FKBP8

MFNG FOXK1

DGKZ

GRK6 VAMP2

RHOXF2 DTX2

RBPJL

COL11A2 LYN

RAC2

HGH1

GET4

DNAJC4 LFNG

MAML3

HEYL

GSK3A

MFNG

PAPPA2

ESR2

CCDC9

BCAN

INPP5D

LFNG CNV/neurodevelopmental gene Signaling pathway gene Connector gene B Average connectivity of CNV genes to C GO term enrichment for connector genes in signaling pathways genes in tissue-specific networks

Hedgehog Notch Wnt en t ns 0.6 0.6 0.6 bo lism ns ub iquitination

* pa ir ne on se to stimulus Signaling on se to stress lle organizationane lle

* an developm * pathway Tissue Org Protein folding Protein Histo Org Signal trans du ction Dopaminergic signaling Axon-dendrite transport Cell cycle re DNA Chromatin organization Protein localization Protein meta Regulation of transcription 0.4 * 0.4 0.4 Resp Resp processing/splicingRNA Brain Hedgehog Heart path length path length path length Kidney

Brain 0.2 0.2 0.2 Notch Heart Kidney 1/shortest 1/shortest 1/shortest Brain Wnt Heart 0.0 0.0 0.0 Kidney KidneyHeart Brain KidneyHeart Brain KidneyHeart Brain Ubiquitous GO enrichments Neuronal GO Non-neuronal enrichments GO enrichments bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

Figure 10

CNV region

( Gene A Gene B Gene C Gene D Gene E Gene F )

Developmental pathways Wnt, Notch, Hedgehog

Biological processes Signal transduction Cell cycle RNA processing Axonal transport Apoptosis Protein degradation

Neuronal phenotypes Non-neuronal phenotypes Autism Cardiac defects Epilepsy Kidney defects Intellectual disability Craniofacial features Micro/macrocephaly Musculoskeletal defects