Chromosome-Centric Human Proteome Project Allies with

Sep 15, 2017 - Here, we present a case study of the role of Y chromosome genes in organ development and how to overcome the challenges facing MPs iden...
1 downloads 15 Views 2MB Size
Subscriber access provided by UOW Library

Review

Chromosome-Centric Human Proteome Project Allies with Developmental Biology: a Case Study of the Role of Y chromosome Genes in Organ Development Anna Meyfour, Paria Pooyan, Sara Pahlavan, Mostafa Rezaei-Tavirani, Hamid Gourabi, Hossein Baharvand, and Ghasem Hosseini Salekdeh J. Proteome Res., Just Accepted Manuscript • DOI: 10.1021/acs.jproteome.7b00446 • Publication Date (Web): 15 Sep 2017 Downloaded from http://pubs.acs.org on September 15, 2017

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

Journal of Proteome Research is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 44

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

Chromosome-Centric Human Proteome Project Allies with Developmental Biology: a Case Study of the Role of Y chromosome Genes in Organ Development

Anna Meyfour1,2, Paria Pooyan1, Sara Pahlavan3, Mostafa Rezaei-Tavirani2, Hamid Gourabi4, Hossein Baharvand3,5*, Ghasem Hosseini Salekdeh1,6*

1

Department of Molecular Systems Biology, Cell Science Research Center, Royan

Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran 2

Proteomics Research Center, Department of Basic Science, Faculty of Paramedical

Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran 3

Department of Stem Cells and Developmental Biology, Cell Science Research Center,

Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran 4

Department of Genetics at Reproductive Biomedicine Research Center, Royan

Institute, Banihashem Sq., Banihashem St., Ressalat highway, Tehran, Iran 5

Department of Developmental Biology, University of Science and Culture, Tehran, Iran

6

Department of Systems Biology, Agricultural Biotechnology Research Institute of Iran,

Karaj, Iran

*Corresponding authors: Ghasem Hosseini Salekdeh, Ph.D., Hossein Baharvand, Ph.D. Royan Institute, Banihashem Sq., Banihashem St., Ressalat highway, Tehran, Iran. Postal Code: 1665659911, P.O. Box: 16635-148, Tel: +98 21 22306485, Fax: +98 21 23562507. Emails: [email protected], [email protected] 1

ACS Paragon Plus Environment

Journal of Proteome Research

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Abstract One of the main goals of Chromosome-Centric Human Proteome Project is to identify protein evidence for missing proteins (MPs). Here, we present a case study of the role of Y chromosome genes in organ development and how to overcome the challenges facing MPs identification by employing human pluripotent stem cell differentiation into cells of different organs yielding unprecedented biological insight into adult silenced proteins. Y chromosome is a male-specific sex chromosome which escapes meiotic recombination. From an evolutionary perspective, Y chromosome has preserved 3% of ancestral genes compared to 98% preservation of the X chromosome based on Ohno’s law. Male specific region of Y chromosome (MSY) contains genes that contribute to central dogma and govern the expression of various targets throughout the genome. One of the most well-known functions of MSY genes are to decide the malespecific characteristics including sex, testis formation and spermatogenesis which are majorly formed by ampliconic gene families. Beyond its role in sexspecific gonad development, MSY genes in co-expression with their X counterparts, as single copy and broadly expressed genes, inhibit haplolethality and play a key role in embryogenesis. The role of X-Y related gene mutations in the development of hereditary syndromes suggests an essential contribution of sex chromosome genes to development. MSY genes, solely and independent of their X counterparts and/or in association with sex hormones, have a considerable impact on fetus development. In this review, we present major recent findings on the contribution of MSY genes to gonad formation, 2

ACS Paragon Plus Environment

Page 2 of 44

Page 3 of 44

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

spermatogenesis, and the brain, heart and kidney development and discuss how Y chromosome proteome project may exploit developmental biology to find missing proteins. Keywords: Chromosome-Centric Human Proteome Project (C-HPP), Y chromosome, gonad formation, organ development

3

ACS Paragon Plus Environment

Journal of Proteome Research

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Introduction The chromosome-centric human proteome project (C-HPP) is designed to address complete map of expression, quantitation, sub-cellular localization, and functional properties of the entire human proteome. One of the main goals of this project is to identify protein evidence for missing proteins (MPs), an inevitably challenging job. The possible limiting factors might reside in I) the expression of MPs in unusual and rare organs or cell types which could be resolved by employing larger entity of samples, II) MPs of the low abundance or unsuitable for mass spectrometry which necessitates the development of more advanced techniques for their identification , III) MPs that are only expressed in specific or early developmental stages and are silent in adult cells 1. The adult silenced genes highlighted the importance of epigenetic regulation as many developmental specific genes become silent due to DNA-histone association and DNA methylation. Therefore, epigenetic manipulation of fully differentiated adult human cells might be a promising approach to discover more MPs involved in development as it has already resulted in identification of 29 MPs specific to spermatogenesis and development 2. Furthermore, another promising approach is to employ novel techniques such as human pluripotent stem cells (hPSCs), which include human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs). hPSCs can selfrenew indefinitely in culture while maintaining the ability to become almost any cell type in the human body.

4

ACS Paragon Plus Environment

Page 4 of 44

Page 5 of 44

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

Here, we present a case study of the role of Y chromosome genes in organ development and introduce hPSC-based developmental studies as a novel potential approach for human proteome project to identify missing proteins which might function during organogenesis and are silent in adult fully differentiated cells. Y chromosome is a haploid, male-specific sex chromosome which escapes meiotic recombination. It has a nonrecombining region (NRY) flanked by two pseudoautosomal regions in either sides which altogether have a 60 Mb size. NRY which is also known as male-specific region of Y chromosome (MSY) comprises 95% of Y chromosome length 3, 4

. According to the last release of neXtProt (2017-0-4, v2.9.0), there are 48 coding

genes on Y chromosome from which, 26 have protein evidence (PE1), 22 do not have experimental protein evidence among which 10 have been validated at transcript level (PE2), 5 are at homology base (PE3) and 7 are at uncertain level (PE5) (www.neXtProt.org)(Table 1). From an evolutionary perspective, X and Y chromosomes are originated from autosomes since 300 million years ago and since then, Y retained only 3% of ancestral genes compared to 98% preservation of the X chromosome

5,

6

. Although Y

chromosome has rapidly lost most of ancestral genes, but remained stable over the last 25 years preserving a particular set of ancestral genes

7, 8

. The reason behind this non-

random preservation was explained in the report by Belllott et al 9. They reconstructed Y chromosome evolution across eight mammals and showed convergent conservancy of X-Y gene pairs in the placental and marsupial lineages, exhibiting long survival despite rapid decay and implicated coherent function in regulation of each central dogma stages. As stated for central dogma, these MSY genes contribute to transcription, 5

ACS Paragon Plus Environment

Journal of Proteome Research

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 6 of 44

translation, and protein stability and in a bigger picture they control the expression of 10, 11

several targets throughout the genome

. The most important mechanisms

underlying their survival are likely to be (1) amplification of multi-copy gene families that govern male gonad development and spermatogenesis

4, 9

, and (2) conservation of

single copies to maintain the correct concentration of extensively-expressed genes vital for both sexes

3, 10, 12

. The X homologue of most dose-sensitive single-copy genes with

breadth of expression escapes X chromosome inactivation

9, 13

and those which are

inactivated have Y homologues with different function. The Turner’s syndrome (monosomy X) is an example of the presence of dose-insufficient sex-related genes. One percent survived fetuses of monosomy X are often mosaic for whole or part of the second sex chromosome (X or Y) 9. Furthermore, some inherent diseases such as Kabuki syndrome, X-linked intellectual disability and autism result from mutations in sex chromosome genes UTX

14

, KDM5C

15

, NLGN4X/NLGN4Y

16

, respectively. The role of

X-Y related gene mutations in the development of these inherent syndromes suggest an essential contribution of sex chromosome genes to embryogenesis. Similarly, PCDH11Y/PCDH11X are sex-linked genes with high expression in the brain and spinal cord during embryonic development

17

and no expression in non-human primates

18

.

Mutations of PCDH11Y/PCDH11X result in disruption of language development and occurrence of non-syndromic speech delay

19

. Y chromosome genes in co-expression

with X homologues can greatly impact the development. X-Y pair genes are expressed during early embryogenesis even before gonad formation

20, 21

. The overexpression of

X-Y pair genes has been identified as early as the onset of zygote gene activation in mammalian development 9. Moreover, sexual dimorphism reportedly emerged in both 6

ACS Paragon Plus Environment

Page 7 of 44

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

genders physiology and pathophysiology particularly the incidence of some hereditary disorders such as autism with 4 times higher rate in males

22

. All these differences from

anatomy to sex-related diseases, originate from sex chromosomes. Additionally, MSY genes, solely and independent of their X counterparts and/or in association with sex hormones, regulate development. These findings necessitates a more focused and detailed study of Y chromosome genes function during development. Our recent findings also showed that Y chromosome genes play an important role during cardiac and nervous system formation

23, 24

. In this review, we discuss the major findings on the

role of Y chromosome genes in various organs development using conventional and novel technologies such as differentiation of hPSCs into multiple cell types of almost all human organs.

Y chromosome in testis formation The phenotypic sex of an individual is mainly determined by the type of gonad that develops in the embryo

25

. Gonad development is a sequential process triggered and

controlled by individual’s sex chromosome complement (specifically the presence or absence of a Y chromosome). This process is followed by the formation of the genital ridge, migration of the primordial germ cells, and the sexual dimorphic differentiation of the gonads

26, 27

. This developmental process directs the bipotential genital ridge to

develop one of the two functionally distinct organs, a testis or an ovary

27

. In human

embryos, formation of the bipotential gonad occurs 33 days post-ovulation (d.p.o.)25. Development of the genital gonads is governed by several central transcription factors that their mutation may inhibit further development of the bipotential gonad 25, 27. Studies 7

ACS Paragon Plus Environment

Journal of Proteome Research

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 8 of 44

on mammalian testis development, identified SRY (a gene located on the short arm of the Y chromosome) as a master gene initiating a cascade of events preceding testis differentiation and ovarian developmental inhibition

25, 28

. An initial SRY expression in

male gonadal ridge is observed during 41-44 d.p.o. that reaches a peak at 44 d.p.o., when sex cords are first visible. SRY expression remains at low levels within the newly formed sex cords throughout the embryonic period and after that

26

. It has been

suggested that SRY may act as a transcription repressor for pluripotency-associated genes and as a transcription activator for differentiation-related genes29. In male genital ridge, SRY expression at its critical level is accompanied by sexually dimorphic expression of its downstream target, SOX9

25

. Compared to SOX9

expression in males, its expression decreases in female developing gonads

30

. SRY

together with steroidogenic factor 1 (SF1, also known as nuclear receptor subfamily 5, group A, member 1 [NR5A1]) directly binds to a specific site (‘TESCO’, testis-specific enhancer of Sox9 core) located upstream of the transcriptional start site of SOX9 between _13 and _10 kb resulting in its upregulation. Upregulated SOX9 autoregulates its expression through binding to TESCO 25, 31. Consequently, the sequential expression of these sex-determining genes causes the activation of downstream targets such as Amh 32, Vnn 33, Pgds and Cbln4 34 (Fig. 1). In addition to activation of Sox9, Sry influences the expression level of various target genes leading to early events of testis differentiation

31

. This male-specific gene takes

part in Sertoli cell fate determination (by binding to and regulating Ptgds) 31, inhibition of ovarian development (by repressing Wnt signaling-dependent gene expression) Sertoli cell proliferation (by targeting Fgf9)

36

35

,

, M germ stem cell niche development (by

8

ACS Paragon Plus Environment

Page 9 of 44

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

targeting Gdnf)

37

, meiotic arrest in germ cells and their M lineage determination (by

activating Cyp26b1)38, and eventual formation of the testis cord and encapsulating the germ cells (by stimulating Wnt5a, Dhh and Ntf3)

31, 39

(Fig. 1). Despite wide range of

target genes and diverse functions of SRY, it is not thoroughly clear how it is being regulated. However, several regulators have been introduced including Gata4, Zfpm240 , Wt134 and Nr5a141, Cbx242 , Jmjd1a, Dax126.

Y chromosome in spermatogenesis Non-obstructive azoospermia in men (complete absence of sperm in the ejaculate) frequently harbours microdeletions within the Azoospermia Factor (AZF) regions (consisting of three sub-regions of AZFa, AZFb and AZFc) of the Y chromosome

43, 44

.

AZFa region of human Y chromosome plays an important role in spermatogenesis as deletions within it, result in irreversible spermatogenic defects causing lack of germ cells in the testis seminiferous tubules diagnosed as Sertoli-cell-only syndrome (SCOS) 45, 46. Further studies demonstrated that deletions within two AZFa genes namely, DDX3Y and USP9Y are responsible for SCOS phenotype role

47

with DDX3Y having a more critical

48

. DDX3Y is a RNA helicase belonging to DEAD-box family which has important

role in cell cycle control, programmed cell death and most of the processes of RNA metabolism including mRNA nuclear export, transcriptional regulation and translational initiation

49

. This gene is expressed in human pre-meiotic spermatogonia during early

stages of fetal testis development

50

(Fig. 1). Additionally, Gueler and colleagues

identified DDX3Y expression after birth in prepubertal Ap spermatogonia suggesting a key role for this protein in male germ cell proliferation and survival 9

ACS Paragon Plus Environment

50

. Furthermore,

Journal of Proteome Research

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 10 of 44

generation of a stable AZFa knockout iPSC line transfected with DDX3Y resulted in formation of prospermatogonial germ cell-like cells as well as germ cell differentiation indicating a regulatory role for DDX3Y in early male germline development 51. AZFb region of Y chromosome contains two main genes (RBMY and PRY) which function during spermatogenesis. While hypospermatogenesis is induced by deletions of all AZFb genes except RBMY and PRY, deletions of these two genes resulted in a complete meiotic arrest

52

. RBMY as a testis-specific splicing factor is expressed at all

stages of germ cell development including A and B spermatogonia, spermatocytes and round spermatids 53(Fig. 1). Its expression and colocalization by splicing factors mediate regulation of RNA splicing at early stages of spermatogenesis

53, 54

. Mice deficient in

Rbmy1 showed spermatogenic arrest which further introduces Rbmy1 as a crucial gene in normal spermatogenesis

55

. Furthermore, recent studies have assigned a role for

sufficient copy numbers of functional RBMY1 in sperm motility which might result in asthenozoospermia in conditions of insufficient copies expression56. PRY gene of AZFb encodes a protein phosphatase that plays a role in regulation of apoptosis required for eliminating non-functional spermatozoa during apoptotic degradation. Although PRY expression was identified in spermatids and spermatozoa, it was absent in premeiotic germ cells (Fig. 1). Therefore, its contribution to meiotic arrest in AZFb deletions is not clear 57. Deleted in Azoospermia (DAZ) genes are known to have crucial role in sperm development

43, 58

. DAZ proteins encoding genes, located within AZFc region, contain a

highly conserved RNA recognition motif (RRM) and a distinctive 24 amino acid sequence known as DAZ repeats

43, 58

. During germ cells differentiation, RNA binding 10

ACS Paragon Plus Environment

Page 11 of 44

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

proteins play crucial roles to store network of mRNAs and control translation timing. It seems that DAZ proteins as RNA binding proteins, take part in translation initiation of mRNA targets

59

. DAZ expression is almost seen at all stages of normal

spermatogenesis from primordial germ cells to spermatogonia, early and late spermatocytes, postmeiotic germ cells and eventually sperms

43

(Fig. 1). DAZ

overexpression resulted in differentiation of both hESCs and iPSCs into primordial germ cells like cells (PGCLCs) and facilitated their further maturation and development

60

.

Furthermore, overexpression of human DAZ in Dazl null mice could partially restore germ cell number

61

. Hence, DAZ proteins may lead to germ cells specification and

meiosis and maintain primordial germ cell population

59

. Chromodomain Y-linked (CDY)

gene of AZFc encodes a protein containing a chromodomain and a histone acetyltransferase catalytic domain essential for generation of protamine-based form of histone-based chromatin structures in late spermatid nuclei spermatogonia, spermatocytes and round spermatids

63

47, 62

. BPY2 is expressed in

(Fig. 1) and believed to interact

with ubiquitin protein ligase E3A which makes it a possible candidate for preservation of human sperm fertility

64

. Y chromosome has copies of TSPY genes which are

expressed in spermatogonia and they regulate the timing of meiosis initiation 65.

Y chromosome in nervous system development Human brain is the most complicated organ, which shows marked sex differences between males and females from anatomical, functional and biochemical perspectives 66

. Sex differences in the brain have been found at all stages of development and can

affect various parameters such as brain area volume, cell number and cytoarchitecture, 11

ACS Paragon Plus Environment

Journal of Proteome Research

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 12 of 44

neural functions, synaptic connectivity, perception, cognition and memory. Moreover, pathophysiology of neurological and neuropsychiatric disorders including the incidence, age of onset, prevalence and severity as well as their symptomatology are sexdependent

67, 68

. Indeed, males and females brains go through different developmental

paths 69. Brain development is a lifelong process that starts prior to the formation of gonads (testis or ovary) and production of sex hormones

69, 70

. Hence, brain’s sexual destiny

starts with the differences in male and female sex chromosomes. Although sex chromosomes account for the differences between the two genders’ brains, the direct roles of their genes are not well studied

66

. As an inevitable consequence, XY and XX

brain cells have distinct transcriptional patterns that dictate their development and function 71. Transcriptional studies revealed the expression of several MSY genes (SRY, RPS4Y1, ZFY, PCDH11Y, TBL1Y, PRKY, USP9Y, DDX3Y, UTY, TMSB4Y, NLGN4Y, HSFY, TXLNGY, KDM5D & EIF1AY) in numerous regions of the male brain at various developmental stages from week 7 post-gestation to adulthood 67, 72, 73. According to the developmental transcriptome data from the BrainSpan atlas (www.brainspan.org), most of the Y chromosome genes are expressed ubiquitously during brain development (Fig. 2). Among these ubiquitously-expressed genes (e.g. RPS4Y1, ZFY, PCDH11Y, TBL1Y, PRKY, USP9Y, DDX3Y, UTY, TMSB4Y, NLGN4Y, CD24P4, TXLNGY, KDM5D & EIF1AY), some keep their fine expression during the entire life (e.g. ZFY, PCDH11Y, TBL1Y, PRKY, USP9Y, DDX3Y, UTY, TMSB4Y, NLGN4Y, TXLNGY, KDM5D & EIF1AY). According to neXtProt, TBL1Y (PE2), PRKY (PE5), TXLNGY & TTTY10 (PE5) & RPS4Y2 (PE2) are all considered as missing proteins. While their gene expression 12

ACS Paragon Plus Environment

Page 13 of 44

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

data during brain development were presented in BrainSpan. Therefore, they can be considered as potential candidates to search for their protein evidence using hPSC derived neural cells which is in accordance with the main goal of Human Proteome Project. RPS4Y1 virtually maintains its sharp expression, prenatal and during infancy; consequently, it may play a role at early stages of brain development. Interestingly, CD24P4 (Known as CD24 in neXtProt), located within MSY region of the Y chromosome, encodes a signal transducer protein and retains the highest gene expression level during the embryonic and prenatal periods followed by a decrease over the rest of life (Fig. 2). However, our understanding of MSY genes function is restricted due to lack of comprehensive studies that should have been done on different periods of brain development. SRY is expressed within various regions of the mammalian male brain especially in dopamine and catecholamine-abundant regions such as the substantia nigra pars compacta (SNpc) and the ventral tegmental area (VTA). SRY protein co-localization with TH (tyrosine hydroxylase)-positive neurons in SNpc and VTA may partly explain the male-specific regulation of dopamine-dependent functions such as the control of movement, attention, and reward-based learning in males

68, 70, 74

. SRY knockdown in

human neuroblastoma cell line (M17) resulted in reduction of enzymes regulating dopamine synthesis and metabolism and consequently dopamine production. However, SRY overexpression caused an increase in enzyme level followed by dopamine synthesis. It has been shown that SRY activates regulatory region of the human TH promoter

74

. These data were in agreement with other studies done in rodents which

indicated SRY conserved function among mammalians 70. 13

ACS Paragon Plus Environment

Journal of Proteome Research

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 14 of 44

Protocadherin 11Y (PCDH11Y) is a human-specific gene emerged on Y chromosome through a duplication and transposition of its paralog, PCDH11X, 6 million years ago

11,

75, 76

. PCDH11Y encodes a protein which consists of an extracellular domain with seven

cadherin repeats, a short transmembrane region, and a cytoplasmic tail

75, 76

. Based on

longitudinal gene expression profiling of males brain, PCDH11Y mRNA is highly expressed in infants while this expression level decreases in childhood and reaches the 72, 73

lowest expression level in adults

. PCDH11Y which is a cell adhesion molecule

takes part in cell survival, fate determination and organization of synapses in different neuronal populations

67

. Johansson and colleagues showed PCDH11Y expression in

NEUN+ neurons, SOX10+ oligodendrocyte precursor cells and ISL1+ motorneurons in different parts of the brain and the central nervous system (CNS)

67

. Blanco and

colleagues showed the upregulation of PCDH11Y in in vitro differentiation of prostate cancer cell lines towards neural lineage, induced by retinoic acid 77. It has been reported that cytoplasmic domain of PCDH11Y interacts with β-catenin resulting in its nuclear accumulation

78

. β-catenin as a component of canonical Wnt signaling pathway is

involved in the formation of the dorso–ventral (D–V) axis in several species such as zebrafish, Xenopus and mice

79, 80

. Wnt signaling is also required for the establishment

of the antero-posterior (A–P), and left–right (L–R) axes of the brain and body

81, 82

.

Interestingly, retinoic acid is also a crucial morphogen involved in the formation of the A–P axis and establishment of the D–V axis 76. NLGN4Y is a Y chromosome specific cell adhesion molecule, a member of neuroliglin (NLGN) family, and a postsynaptic transmembrane protein that interacts with neurexin (NRXN)83. Like other members of this family, NLGN4Y acts as an inducer and 14

ACS Paragon Plus Environment

Page 15 of 44

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

modulator of synaptic activity. NLGN4Y regulates synaptic adhesion and development through its extracellular domain which is an acetylcholinesterase (AChE) homologue without enzymatic activity

83, 84

. Like other cell adhesion molecules, therefore, NLGN4Y

is an essential factor for formation of functional synapses and its mutations as well as translation or function failure, may cause neurological disorders, such as autism. It has been shown that both NLGN4Y mutation in XY and overexpression in XYY, are associated with prominent risk of autism

85-87

. Another interesting finding on NLGN4Y

was that its expression reduced during brain development from early postconception weeks to adulthood 67, 72. In a study designed to address Y chromosome human proteome project, Vakilian and colleagues profiled the expression of 23 MSY genes and 15 of their X-linked homologues during neural cell differentiation of a human embryonic carcinoma cell line, NTRA-2. The expression of RBMY1, EIF1AY, DDX3Y, HSFY1, BPY2, PCDH11Y, UTY, RPS4Y1, USP9Y, SRY, PRY and ZFY were significantly increased during differentiation.

23

. Furthermore, DDX3Y knockdown in neural differentiation of NTRA-2

resulted in cell growth arrest at G1/S phase and overexpression of pro-apoptotic proteins 23.

Y chromosome in cardiac development Gender-related differences in basal cardiovascular function have been reported for two decades. There are physiological differences in left ventricular ejection fraction (LVEF) and stroke volume (SV), resulting in specific heart rates in men compared to women 89

88,

. These sex-related differences have been also observed at transcriptome level in 15

ACS Paragon Plus Environment

Journal of Proteome Research

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 16 of 44

human and rodents, many of which such as USP9Y, RPS4Y1, DDX3Y, XIST, TIMP1, UTY, ZFY and PRKY, are expressed on X-Y chromosomes

90

. Interestingly, sex

dimorphism has been observed in mitochondria transcriptome of rats heart as well

91

.

Moreover, there are gender-related differences in the prevalence of cardiovascular diseases

such

as

atherosclerosis,

myocardial

infarction,

myocarditis,

dilated

cardiomyopathy and heart failure with higher rates in men compared to hypertention in women

92-94

. Molecular biology studies showed that these sexual disparities present at

transcriptional level particularly in the expression of Y chromosome genes such as DDX3Y, RPS4Y1, USP9Y and JARID1D

95

. Therefore, sex can have an impact on

regulatory mechanisms of cardiovascular physiology and pathophysiology. While female protection may be in part due to estrogen influence

96

and male susceptibility due to

testicular hormone 97, but Y chromosome seems to play an important role in these malespecific cardiac functions

98

. A growing body of evidence has been emerged on Y

chromosome function by crossing Y chromosomes from different mouse strains onto a single background strain showing its impact on cardiac phenotype

99

. Llamas et al

exchanged the Y chromosome of C57BL (ChrYC57) with that of A/J mice (ChrYA), resulting in consomic strains (C57.YA and A/J.YC57)

100

. While both mice strains are

normotensive, C57BL mice hearts developed an eccentric hypertrophy phenotype compared to A/J mice

101

. Surprisingly, the cardiomyocytes size appeared to be smaller

in C57.YA compared to C57BL. On the other hand, A/J.YC57 obtained larger cardiomyocytes in comparison to A/J mice, indicating a contributory role for Y chromosome in the loci governing cardiomyocytes size in male mice. These findings provided valuable hints on Y chromosome genes contribution to cardiovascular 16

ACS Paragon Plus Environment

Page 17 of 44

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

phenotype. To further confirm this hypothesis, they studied the cardiomyocyte’s phenotype in consomic strains undergone pre-pubertal castration

102

. The strains

carrying YC57 showed a decrease in cardiomyocytes size in the absence of post-pubertal testosterone. However, YA failed to induce changes in cardiomyocytes size suggesting their insensitivity to the hypertrophic effects of post-pubertal testosterone. However, the transcriptome analyses showed different expression level for many cardiac genes in unaffected C57.YA cardiomyocytes even greater than that observed in C57BL mice. Furthermore, the cardiac transcriptome of castrated C57BL mice was compared to their sham-operated counterpart which showed a different expression of cardiac genes independent of testosterone

102

. These findings show the role of Y chromosome in

cardiac cell properties in the presence or absence of androgen regulations. To better identify how Y chromosome polymorphisms may impact cardiac characteristics, they further focused on these two male mice strains, C57BL and C57.YA103 and found different distribution of androgen receptors (ARs) in chromatins from neonatal male hearts of these two strains. Furthermore, they identified genes with AR occupancy which were enriched in biological processes related to heart development as well as hypertrophy. They also showed different chromatin accessibility and strain-specific H3K4me3-marked genes. Moreover, when they profiled Y chromosome genes in the heart of these two strains, they observed different expression of Jarid1d, Uty, Ddx3y and Eif2s3y among which Jarid1d and Uty displayed similar pattern in both heart and testis in a narrow window of 24 h after birth. These data, for the first time, provided a conceptual framework to unravel how Y chromosome regulates cardiac phenotype by affecting chromatin accessibility and neonatal programming 17

ACS Paragon Plus Environment

103

.

Androgens affect

Journal of Proteome Research

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 18 of 44

cardiac circadian rhythm, expression of contractile genes and myocardial functional characteristics by influencing MSY genetic materials in the first 24 h of neonatal life. These findings introduce androgens as a necessary but not sufficient factor to some particular cardiac functions. Altogether, they suggested MSY as the origin of malespecific genetic factors which can modulate the androgen functions among individuals 98

.

As described previously, UTY was identified as one of MSY genes which may function in cardiac development. UTY and its X counterpart have been shown to be overexpressed during cardiac differentiation of ESCs

104

. These data identified a key

role for UTX in cardiac lineage differentiation of ESCs. UTX∆/∆ female mice died at embryonic stage; however, UTX∆/y male mice displayed various phenotypes including sever cardiac developmental defects suggesting a partial compensation of UTY for its X homologue. Furthermore, UTX-null mice and UTX-null ESCs provided information on UTX function as a coactivator of core cardiac transcription factors

104

. Another study

showed that male UTX knockout (KO) with UTY expression survived until birth, while female UTX KO embryos died from developmental defects in mesodermal lineages such as mesoderm-derived posterior notochord, cardiac and hematopoietic tissues. These results suggested a role for UTX/UTY in early embryonic development

105, 106

.A

recent study showed that UTX knockdown inhibited the direct cardiac reprogramming ability of a miR combo (miR-1, miR-133, miR-208 and miR-499) by restoring the level of H3K27me3 in fibroblasts 107. To better understand the Y chromosome function in cardiac development, the hormonefree models should be used. To address Y chromosome human proteome project, our 18

ACS Paragon Plus Environment

Page 19 of 44

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

group performed the first whole expression profiling of Y chromosome genes in a cellbased model of in vitro cardiac differentiation by using male hESCs. We identified alterations in gene expression of TBL1Y, PCDH11Y, ZFY, KDM5D, USP9Y, RPS4Y, DDX3Y, PRY, XKRY, BCORP1, RBMY, HSFY and UTY (Fig. 3) which influenced protein expression and localization during cardiac differentiation. Among these, PCDH11Y, ZFY, KDM5D, USP9Y, RPS4Y, DDX3Y, UTY, TBL1Y, PRY and BCORP1 showed overexpression at cardiac mesoderm development. Some other MSY genes such as RBMY, HSFY and BCORP1 were overexpressed during late cardiogenesis. All MSY genes which were expressed and showed alterations during cardiac differentiation of hESCs, are summarized in figure 3. Because TBL1Y overexpression appeared in a thoroughly different pattern compared to its X homologue24, we further studied the function of this missing protein in cardiac differentiation of hESCs. TBL1Y knock down negatively affected cardiac differentiation by stabilization of CtBP as a member of Notch co-repressor complex resulting in inhibition of Notch signaling activation24. Perissi and colleagues showed that Notch signaling activation requires the release of a dual repression caused by distinct corepressor complexes, CtBP and NCoR/SMRT

108

.

TBL1X/TBL1Y and TBLR1 are F box/WD40 containing factors that function as exchangers in the release of dual repression by dismissal of CtBP and NCoR/SMART from corepressor complex resulting in transcriptional activation of Notch target genes such as Hes124

108

(Fig. 4). Moreover, a delay in the onset of beating and an abnormal

contraction pattern was observed in cardiomyocytes lacking TBL1Y24 . Other reports also illustrated cardiac developmental defects in patients carrying TBL1Y mutations resulting in non-syndromic coarctation

109

. Furthermore, UTY overexpression during in 19

ACS Paragon Plus Environment

Journal of Proteome Research

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 20 of 44

vitro cardiac differentiation24 was in agreement with previous studies reporting UTY function in cardiac development

104-107

. Moreover, OFCD patients carrying BCOR

mutations show developmental heart defects with majority having septal malformations 110

. A growing body of evidence show a key role for BCOR as a transcriptional regulator

during early embryogenesis specifically cardiogenesis BCORP1

identification

at

transcript

level

and

111

its

. Based on our findings of overrepresentation

during

cardiogenesis, it may also impact cardiac development which has to be further investigated. These findings suggest that MSY genes either solely or in cooperation with their X homologue, regulate cardiogenesis. Therefore, there is an inevitable need for new experimental paradigm to increase the foundation of basic knowledge on MSY genes expression and their impact on gender-related developmental differences during cardiogenesis. Cardiogenic differentiation of hPSCs might be a possible approach for such cardiac developmental studies as well as congenital cardiovascular disorders.

Y chromosome in kidney development The kidney is an organ which filters wastes from body fluids, regulates blood electrolytes and maintains whole-body homeostasis. A key function of kidney is to regulate blood pressure by activation of renin-angiotensin system. Gender differences were described to affect the morphology and function of kidneys

112

. Despite similar

number of nephrons in both females and males, the mass of kidney is higher in the latter 113. Furthermore, it has been reported that the composition of renal transporters as well as infiltration of specific organic compounds were gender-related

112

. These sex-

related differences were reflected in the higher prevalence and progression of renal 20

ACS Paragon Plus Environment

Page 21 of 44

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

diseases in males as shown by 50% more men diagnosed with end stage renal disease (ESRD)

114

. Sex hormones might impact the progression of renal diseases in females

and males resulting in estradiol-related protection and testosterone-associated vulnerability, respectively

115

. However, these sex dimorphism might be also mediated

by the expression of sex chromosomal genes

116

. The gene expression of healthy

glomeruli and tubuli showed sexual dimorphism with enriched sex chromosome genes such as EHD2, DDX3Y, EIF1AY, CYorf14, ESPL1, KDM5D, PNPLa4, RPS4Y1 and XIST

117

. Sexual dimorphism of renal gene expression was also reported in ESRD

particularly the following nine genes: XIST, DDX3Y, KDM5D, CYorf14, EIF1AY, HDHD1A, RPS4Y1, STS and ZRSR2

117

. Moreover, renin-angiotensin system (RAS)

showed gender-related differences resulting in sex-specific blood-pressure regulation 118, 119

. Consomic rat models provided the first evidence on the contribution of Y

chromosome to regulation of blood pressure

120

. SRY was determined as a Y

chromosome gene responsible for elevation of the blood pressure by affecting RAS

121,

122

. SRY and its X homologue, SOX3, reportedly regulate renin and angiotensinogen 123

promoters

. Moreover, SRY expression/SOX3 absence in the kidney resulted in a

male-specific regulation of renin production. Therefore, SRY/SOX3-mediated regulation of RAS is a rate–limiting step in the kidney (due to SRY expression in kidney) resulting in a male-specific control of blood pressure 124. The overexpression of RAS components has been shown during kidney development. Furthermore, transgenic mice with targeted inactivation of renin kidney

125

or angiotensin

126

showed developmental defects in the

127

. Based on these findings, SRY might function in kidney development by

activation of SRY-mediated renin/angiotensin promoters. 21

ACS Paragon Plus Environment

Journal of Proteome Research

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 22 of 44

PRKX gene expression was identified in the fetal kidney as well as polycystic kidney, though it is absent in adult healthy kidney

128

. PRKX overexpression was shown to be

able to activate cellular migration and epithelial tubular formation in vitro

128

as well as

ureteric bud branching and glomeruli morphogenesis in embryonic mouse kidney explants suggesting its possible role in kidney development

129, 130

. Constitutive PRKX

activation in human autosomal dominant polycystic kidney disease (ADPKD) epithelial cell lines has shown to rescue adhesion and migration defects that could compensate PKD1 mutation in ADPKD phenotype

131

. Because PRKX escapes X inactivation,

therefore, it requires the expression of both alleles to exert its normal function. Furthermore, it has a highly similar sequence with its Y counterpart. Therefore, we suggest that PRKY might play an important role in male kidney development as well.

Conclusion and perspective Y chromosome governs male-specific characteristics including sex determination, testis formation and spermatogenesis. Most of Y chromosome genes contribute to these distinct male-specific functions particularly through ampliconic gene families (Table 1). Beyond its role in sex-specific gonad development, MSY genes in co-operation with their X counterparts as single copy and broadly-expressed genes prevent haplolethality and play a key role in fetus development. Moreover, Y chromosome genes can exhibit unique functions causing sexual dimorphism, independent or in association with sex hormones, dissimilar to their X-linked collective tasks during organ development. However, little knowledge exists on MSY proteins and their role during different organs development (Table 1). This might have been originated from the biological limitations 22

ACS Paragon Plus Environment

Page 23 of 44

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

and/or technical issues such as expression of MSY proteins in rare organs or cell types, low abundance of proteins, expression in specific or early developmental stages and being silent in adult tissues and cells, high similarity with X counterparts, and lack of specific antibodies again MSY proteins. In Y chromosome Human Proteome Project, we aimed to overcome these obstacles to unravel the gene expression and protein function of Y chromosome genes during development, taking the advantage of hPSCs. We aim to identify the existence, abundance and localization of MSY proteins during organ development specially MPs that are only expressed in early developmental stages and are silent in adult cells. Two unique characteristics of hPSCs make them well suited for these studies. First, hPSCs have unlimited self-renewal capacity, providing abundant material for mass spectrometry analysis. The discovery of patient-specific induced pluripotent stem cells (iPSCs) paved the way for developmental studies as well as end-stage pathophysiology of hereditary disorders and consequently, discovery of novel therapeutic. The access to the patient-specific iPSCs may also facilitate the discovery of missing proteins. Second, hPSCs have the potential to generate every adult cell type, offering an attractive window into proteins expressed during human development. Their in vitro culture system also provides a rapid, cost-effective way to interrogate the function of a gene during a specific developmental process and to identify highly similar missing proteins. Proteins such as RPS4Y2 and TBL1Y have been designated as “marginally distinguished” in PeptideAtlas due to their very high similar to their X homologue 23

ACS Paragon Plus Environment

Journal of Proteome Research

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 24 of 44

counterpart. Therefore, there is no credible evidence to suggest that they can be distinguished from their X counterparts with the mass spectrometry data. Families of proteins with high similarity represent many of the indistinguishable and marginally distinguished matches in the databases.

They may remain unidentifiable, unless

uniquely-mapping or proteotypic peptides can be found to differentiate them 132. We recently exploited a genomics tool as a complement to protein studies to find TBL1Y, missing protein of Y chromosome24. We generated TBL1Y antibody using a peptide and showed that while TBL1Y expressed sharply in XY hESCs, no expression could be detected in XX hESCs. Furthermore, we observed that TBL1Y also showed overexpression and a thoroughly different expression pattern compared to its Xhomologue during differentiation of hESCs to cardiomyocyte. Using siRNA approach, we demonstrated that the RNA and protein abundance of TBL1Y declined in the siRNA transfected cells. All together, we could present compelling experimental evidence to distinguish TBL1Y from TBL1X, and propose reclassification of TBL1Y as ‘found missing protein’ (PE1). We expect that several Y chromosome missing proteins will be identified by mass spectrometry analysis of hPSC derived cells. Alternatively, in vitro culture system of hPSC can also provide a unique platform to identify the highly similar proteins and to interrogate their functions using various genomics and proteomics tools. The hPSCs, therefore, can act as a strong complement to C-HPP.

24

ACS Paragon Plus Environment

Page 25 of 44

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

Acknowledgment The Human Y Chromosome Proteome Project is supported by a grant from Royan Institute. Conflict of interest: The authors declare no competing financial interest.

25

ACS Paragon Plus Environment

Journal of Proteome Research

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 26 of 44

References 1. Horvatovich, P.; Lundberg, E. K.; Chen, Y.-J.; Sung, T.-Y.; He, F.; Nice, E. C.; Goode, R. J.; Yu, S.; Ranganathan, S.; Baker, M. S., Quest for missing proteins: update 2015 on chromosome-centric human proteome project. Journal of proteome research 2015, 14, (9), 3415-3431. 2. Yang, L.; Lian, X.; Zhang, W.; Guo, J.; Wang, Q.; Li, Y.; Chen, Y.; Yin, X.; Yang, P.; Lan, F.; He, Q. Y.; Zhang, G.; Wang, T., Finding Missing Proteins from the Epigenetically Manipulated Human Cell with Stringent Quality Criteria. J Proteome Res 2015, 14, (9), 3645-57. 3. Skaletsky, H.; Kuroda-Kawaguchi, T.; Minx, P. J.; Cordum, H. S.; Hillier, L.; Brown, L. G.; Repping, S.; Pyntikova, T.; Ali, J.; Bieri, T.; Chinwalla, A.; Delehaunty, A.; Delehaunty, K.; Du, H.; Fewell, G.; Fulton, L.; Fulton, R.; Graves, T.; Hou, S. F.; Latrielle, P.; Leonard, S.; Mardis, E.; Maupin, R.; McPherson, J.; Miner, T.; Nash, W.; Nguyen, C.; Ozersky, P.; Pepin, K.; Rock, S.; Rohlfing, T.; Scott, K.; Schultz, B.; Strong, C.; Tin-Wollam, A.; Yang, S. P.; Waterston, R. H.; Wilson, R. K.; Rozen, S.; Page, D. C., The male-specific region of the human Y chromosome is a mosaic of discrete sequence classes. Nature 2003, 423, (6942), 825-37. 4. Jangravi, Z.; Alikhani, M.; Arefnezhad, B.; Sharifi Tabar, M.; Taleahmad, S.; Karamzadeh, R.; Jadaliha, M.; Mousavi, S. A.; Ahmadi Rastegar, D.; Parsamatin, P.; Vakilian, H.; Mirshahvaladi, S.; Sabbaghian, M.; Mohseni Meybodi, A.; Mirzaei, M.; Shahhoseini, M.; Ebrahimi, M.; Piryaei, A.; Moosavi-Movahedi, A. A.; Haynes, P. A.; Goodchild, A. K.; Nasr-Esfahani, M. H.; Jabbari, E.; Baharvand, H.; Sedighi Gilani, M. A.; Gourabi, H.; Salekdeh, G. H., A fresh look at the male-specific region of the human Y chromosome. J Proteome Res 2013, 12, (1), 6-22. 5. Bellott, D. W.; Skaletsky, H.; Pyntikova, T.; Mardis, E. R.; Graves, T.; Kremitzki, C.; Brown, L. G.; Rozen, S.; Warren, W. C.; Wilson, R. K.; Page, D. C., Convergent evolution of chicken Z and human X chromosomes by expansion and gene acquisition. Nature 2010, 466, (7306), 612-6. 6. Mueller, J. L.; Skaletsky, H.; Brown, L. G.; Zaghlul, S.; Rock, S.; Graves, T.; Auger, K.; Warren, W. C.; Wilson, R. K.; Page, D. C., Independent specialization of the human and mouse X chromosomes for the male germ line. Nat Genet 2013, 45, (9), 1083-7. 7. Hughes, J. F.; Skaletsky, H.; Pyntikova, T.; Minx, P. J.; Graves, T.; Rozen, S.; Wilson, R. K.; Page, D. C., Conservation of Y-linked genes during human evolution revealed by comparative sequencing in chimpanzee. Nature 2005, 437, (7055), 100-3. 8. Hughes, J. F.; Skaletsky, H.; Brown, L. G.; Pyntikova, T.; Graves, T.; Fulton, R. S.; Dugan, S.; Ding, Y.; Buhay, C. J.; Kremitzki, C.; Wang, Q.; Shen, H.; Holder, M.; Villasana, D.; Nazareth, L. V.; Cree, A.; Courtney, L.; Veizer, J.; Kotkiewicz, H.; Cho, T. J.; Koutseva, N.; Rozen, S.; Muzny, D. M.; Warren, W. C.; Gibbs, R. A.; Wilson, R. K.; Page, D. C., Strict evolutionary conservation followed rapid gene loss on human and rhesus Y chromosomes. Nature 2012, 483, (7387), 82-6. 9. Bellott, D. W.; Hughes, J. F.; Skaletsky, H.; Brown, L. G.; Pyntikova, T.; Cho, T. J.; Koutseva, N.; Zaghlul, S.; Graves, T.; Rock, S.; Kremitzki, C.; Fulton, R. S.; Dugan, S.; Ding, Y.; Morton, D.; Khan, Z.; Lewis, L.; Buhay, C.; Wang, Q.; Watt, J.; Holder, M.; Lee, S.; Nazareth, L.; Alfoldi, J.; Rozen, S.; Muzny, D. M.; Warren, W. C.; Gibbs, R. A.; 26

ACS Paragon Plus Environment

Page 27 of 44

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

Wilson, R. K.; Page, D. C., Mammalian Y chromosomes retain widely expressed dosage-sensitive regulators. Nature 2014, 508, (7497), 494-9. 10. Lahn, B. T.; Page, D. C., Functional coherence of the human Y chromosome. Science 1997, 278, (5338), 675-80. 11. Cortez, D.; Marin, R.; Toledo-Flores, D.; Froidevaux, L.; Liechti, A.; Waters, P. D.; Grutzner, F.; Kaessmann, H., Origins and functional evolution of Y chromosomes across mammals. Nature 2014, 508, (7497), 488-93. 12. Kaiser, V. B.; Zhou, Q.; Bachtrog, D., Nonrandom gene loss from the Drosophila miranda neo-Y chromosome. Genome Biol Evol 2011, 3, 1329-37. 13. Carrel, L.; Willard, H. F., X-inactivation profile reveals extensive variability in Xlinked gene expression in females. Nature 2005, 434, (7031), 400-4. 14. Bogershausen, N.; Gatinois, V.; Riehmer, V.; Kayserili, H.; Becker, J.; Thoenes, M.; Simsek-Kiper, P. O.; Barat-Houari, M.; Elcioglu, N. H.; Wieczorek, D.; Tinschert, S.; Sarrabay, G.; Strom, T. M.; Fabre, A.; Baynam, G.; Sanchez, E.; Nurnberg, G.; Altunoglu, U.; Capri, Y.; Isidor, B.; Lacombe, D.; Corsini, C.; Cormier-Daire, V.; Sanlaville, D.; Giuliano, F.; Le Quan Sang, K. H.; Kayirangwa, H.; Nurnberg, P.; Meitinger, T.; Boduroglu, K.; Zoll, B.; Lyonnet, S.; Tzschach, A.; Verloes, A.; Di Donato, N.; Touitou, I.; Netzer, C.; Li, Y.; Genevieve, D.; Yigit, G.; Wollnik, B., Mutation Update for Kabuki Syndrome Genes KMT2D and KDM6A and Further Delineation of X-Linked Kabuki Syndrome Subtype 2. Hum Mutat 2016, 37, (9), 847-64. 15. Grafodatskaya, D.; Chung, B. H.; Butcher, D. T.; Turinsky, A. L.; Goodman, S. J.; Choufani, S.; Chen, Y. A.; Lou, Y.; Zhao, C.; Rajendram, R.; Abidi, F. E.; Skinner, C.; Stavropoulos, J.; Bondy, C. A.; Hamilton, J.; Wodak, S.; Scherer, S. W.; Schwartz, C. E.; Weksberg, R., Multilocus loss of DNA methylation in individuals with mutations in the histone H3 lysine 4 demethylase KDM5C. BMC Med Genomics 2013, 6, 1. 16. Serajee, F. J.; Mahbubul Huq, A. H., Association of Y chromosome haplotypes with autism. J Child Neurol 2009, 24, (10), 1258-61. 17. Yoshida, K.; Sugano, S., Identification of a novel protocadherin gene (PCDH11) on the human XY homology region in Xq21.3. Genomics 1999, 62, (3), 540-3. 18. Wilson, N. D.; Ross, L. J.; Crow, T. J.; Volpi, E. V., PCDH11 is X/Y homologous in Homo sapiens but not in Gorilla gorilla and Pan troglodytes. Cytogenet Genome Res 2006, 114, (2), 137-9. 19. Speevak, M. D.; Farrell, S. A., Non-syndromic language delay in a child with disruption in the Protocadherin11X/Y gene pair. Am J Med Genet B Neuropsychiatr Genet 2011, 156B, (4), 484-9. 20. Dewing, P.; Shi, T.; Horvath, S.; Vilain, E., Sexually dimorphic gene expression in mouse brain precedes gonadal differentiation. Brain Res Mol Brain Res 2003, 118, (12), 82-90. 21. Arnold, A. P., Sex chromosomes and brain gender. Nat Rev Neurosci 2004, 5, (9), 701-8. 22. Skuse, D. H., Imprinting, the X-chromosome, and the male brain: explaining sex differences in the liability to autism. Pediatr Res 2000, 47, (1), 9-16. 23. Vakilian, H.; Mirzaei, M.; Sharifi Tabar, M.; Pooyan, P.; Habibi Rezaee, L.; Parker, L.; Haynes, P. A.; Gourabi, H.; Baharvand, H.; Salekdeh, G. H., DDX3Y, a Male-Specific Region of Y Chromosome Gene, May Modulate Neuronal Differentiation. J Proteome Res 2015, 14, (9), 3474-83. 27

ACS Paragon Plus Environment

Journal of Proteome Research

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 28 of 44

24. Meyfour, A.; Ansari, H.; Pahlavan, S.; Mirshahvaladi, S.; Rezaei-Tavirani, M.; Gourabi, H.; Baharvand, H.; Salekdeh, G. H., Y Chromosome Missing Protein, TBL1Y, May Play an Important Role in Cardiac Differentiation. Journal of Proteome Research 2017, 10.1021/acs.jproteome.7b00391. 25. Eggers, S.; Ohnesorg, T.; Sinclair, A., Genetic regulation of mammalian gonad development. Nat Rev Endocrinol 2014, 10, (11), 673-83. 26. Lucas-Herald, A. K.; Bashamboo, A., Gonadal development. Endocr Dev 2014, 27, 1-16. 27. She, Z. Y.; Yang, W. X., Molecular mechanisms involved in mammalian primary sex determination. J Mol Endocrinol 2014, 53, (1), R21-37. 28. Kashimada, K.; Koopman, P., Sry: the master switch in mammalian sex determination. Development 2010, 137, (23), 3921-30. 29. Kakhki, S. A.; Shahhoseini, M.; Salekdeh, G. H., Comparative SRY incorporation on the regulatory regions of pluripotency/differentiation genes in human embryonic carcinoma cells after retinoic acid induction. Molecular and cellular biochemistry 2013, 376, (1-2), 145-150. 30. Sekido, R.; Bar, I.; Narvaez, V.; Penny, G.; Lovell-Badge, R., SOX9 is upregulated by the transient expression of SRY specifically in Sertoli cell precursors. Dev Biol 2004, 274, (2), 271-9. 31. Li, Y.; Zheng, M.; Lau, Y. F., The sex-determining factors SRY and SOX9 regulate similar target genes and promote testis cord formation during testicular differentiation. Cell Rep 2014, 8, (3), 723-33. 32. Arango, N. A.; Lovell-Badge, R.; Behringer, R. R., Targeted mutagenesis of the endogenous mouse Mis gene promoter: in vivo definition of genetic pathways of vertebrate sexual development. Cell 1999, 99, (4), 409-19. 33. Wilson, M. J.; Jeyasuria, P.; Parker, K. L.; Koopman, P., The transcription factors steroidogenic factor-1 and SOX9 regulate expression of Vanin-1 during mouse testis development. J Biol Chem 2005, 280, (7), 5917-23. 34. Bradford, S. T.; Wilhelm, D.; Bandiera, R.; Vidal, V.; Schedl, A.; Koopman, P., A cell-autonomous role for WT1 in regulating Sry in vivo. Hum Mol Genet 2009, 18, (18), 3429-38. 35. Lau, Y. F.; Li, Y., The human and mouse sex-determining SRY genes repress the Rspol/beta-catenin signaling. J Genet Genomics 2009, 36, (4), 193-202. 36. Colvin, J. S.; Green, R. P.; Schmahl, J.; Capel, B.; Ornitz, D. M., Male-to-female sex reversal in mice lacking fibroblast growth factor 9. Cell 2001, 104, (6), 875-89. 37. Oatley, J. M.; Brinster, R. L., Regulation of spermatogonial stem cell self-renewal in mammals. Annu Rev Cell Dev Biol 2008, 24, 263-86. 38. Bowles, J.; Knight, D.; Smith, C.; Wilhelm, D.; Richman, J.; Mamiya, S.; Yashiro, K.; Chawengsaksophak, K.; Wilson, M. J.; Rossant, J.; Hamada, H.; Koopman, P., Retinoid signaling determines germ cell fate in mice. Science 2006, 312, (5773), 596600. 39. Clement, T. M.; Bhandari, R. K.; Sadler-Riggleman, I.; Skinner, M. K., SRY directly regulates the neurotrophin 3 promoter during male sex determination and testis development in rats. Biol Reprod 2011, 85, (2), 277-84. 40. Tevosian, S. G.; Albrecht, K. H.; Crispino, J. D.; Fujiwara, Y.; Eicher, E. M.; Orkin, S. H., Gonadal differentiation, sex determination and normal Sry expression in 28

ACS Paragon Plus Environment

Page 29 of 44

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

mice require direct interaction between transcription partners GATA4 and FOG2. Development 2002, 129, (19), 4627-34. 41. de Santa Barbara, P.; Mejean, C.; Moniot, B.; Malcles, M. H.; Berta, P.; BoizetBonhoure, B., Steroidogenic factor-1 contributes to the cyclic-adenosine monophosphate down-regulation of human SRY gene expression. Biol Reprod 2001, 64, (3), 775-83. 42. Katoh-Fukui, Y.; Miyabayashi, K.; Komatsu, T.; Owaki, A.; Baba, T.; Shima, Y.; Kidokoro, T.; Kanai, Y.; Schedl, A.; Wilhelm, D.; Koopman, P.; Okuno, Y.; Morohashi, K., Cbx2, a polycomb group gene, is required for Sry gene expression in mice. Endocrinology 2012, 153, (2), 913-24. 43. Vangompel, M. J.; Xu, E. Y., The roles of the DAZ family in spermatogenesis: More than just translation? Spermatogenesis 2011, 1, (1), 36-46. 44. Carrell, D. T.; De Jonge, C.; Lamb, D. J., The genetics of male infertility: a field of study whose time is now. Arch Androl 2006, 52, (4), 269-74. 45. Kamp, C.; Huellen, K.; Fernandes, S.; Sousa, M.; Schlegel, P. N.; Mielnik, A.; Kleiman, S.; Yavetz, H.; Krause, W.; Kupker, W.; Johannisson, R.; Schulze, W.; Weidner, W.; Barros, A.; Vogt, P. H., High deletion frequency of the complete AZFa sequence in men with Sertoli-cell-only syndrome. Mol Hum Reprod 2001, 7, (10), 98794. 46. Ahmadi Rastegar, D.; Sharifi Tabar, M.; Alikhani, M.; Parsamatin, P.; Sahraneshin Samani, F.; Sabbaghian, M.; Sadighi Gilani, M. A.; Mohammad Ahadi, A.; Mohseni Meybodi, A.; Piryaei, A., Isoform-Level Gene Expression Profiles of Human Y Chromosome Azoospermia Factor Genes and Their X Chromosome Paralogs in the Testicular Tissue of Non-Obstructive Azoospermia Patients. Journal of proteome research 2015, 14, (9), 3595-3605. 47. Vogt, P. H., Azoospermia factor (AZF) in Yq11: towards a molecular understanding of its function for human male fertility and spermatogenesis. Reprod Biomed Online 2005, 10, (1), 81-93. 48. Krausz, C.; Degl'Innocenti, S.; Nuti, F.; Morelli, A.; Felici, F.; Sansone, M.; Varriale, G.; Forti, G., Natural transmission of USP9Y gene mutations: a new perspective on the role of AZFa genes in male fertility. Hum Mol Genet 2006, 15, (18), 2673-81. 49. Linder, P.; Jankowsky, E., From unwinding to clamping - the DEAD box RNA helicase family. Nat Rev Mol Cell Biol 2011, 12, (8), 505-16. 50. Gueler, B.; Sonne, S. B.; Zimmer, J.; Hilscher, B.; Hilscher, W.; Graem, N.; Rajpert-De Meyts, E.; Vogt, P. H., AZFa protein DDX3Y is differentially expressed in human male germ cells during development and in testicular tumours: new evidence for phenotypic plasticity of germ cells. Hum Reprod 2012, 27, (6), 1547-55. 51. Ramathal, C.; Angulo, B.; Sukhwani, M.; Cui, J.; Durruthy-Durruthy, J.; Fang, F.; Schanes, P.; Turek, P. J.; Orwig, K. E.; Reijo Pera, R., DDX3Y gene rescue of a Y chromosome AZFa deletion restores germ cell formation and transcriptional programs. Sci Rep 2015, 5, 15041. 52. Ferlin, A.; Moro, E.; Rossi, A.; Dallapiccola, B.; Foresta, C., The human Y chromosome's azoospermia factor b (AZFb) region: sequence, structure, and deletion analysis in infertile men. J Med Genet 2003, 40, (1), 18-24. 29

ACS Paragon Plus Environment

Journal of Proteome Research

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 30 of 44

53. Elliott, D. J.; Oghene, K.; Makarov, G.; Makarova, O.; Hargreave, T. B.; Chandley, A. C.; Eperon, I. C.; Cooke, H. J., Dynamic changes in the subnuclear organisation of pre-mRNA splicing proteins and RBM during human germ cell development. J Cell Sci 1998, 111 ( Pt 9), 1255-65. 54. Dreumont, N.; Bourgeois, C. F.; Lejeune, F.; Liu, Y.; Ehrmann, I. E.; Elliott, D. J.; Stevenin, J., Human RBMY regulates germline-specific splicing events by modulating the function of the serine/arginine-rich proteins 9G8 and Tra2-{beta}. J Cell Sci 2010, 123, (Pt 1), 40-50. 55. Mahadevaiah, S. K.; Odorisio, T.; Elliott, D. J.; Rattigan, A.; Szot, M.; Laval, S. H.; Washburn, L. L.; McCarrey, J. R.; Cattanach, B. M.; Lovell-Badge, R.; Burgoyne, P. S., Mouse homologues of the human AZF candidate gene RBM are expressed in spermatogonia and spermatids, and map to a Y chromosome deletion interval associated with a high incidence of sperm abnormalities. Hum Mol Genet 1998, 7, (4), 715-27. 56. Yan, Y.; Yang, X.; Liu, Y.; Shen, Y.; Tu, W.; Dong, Q.; Yang, D.; Ma, Y.; Yang, Y., Copy number variation of functional RBMY1 is associated with sperm motility: an azoospermia factor-linked candidate for asthenozoospermia. Human Reproduction 2017, 32, (7), 1-11. 57. Stouffs, K.; Lissens, W.; Verheyen, G.; Van Landuyt, L.; Goossens, A.; Tournaye, H.; Van Steirteghem, A.; Liebaers, I., Expression pattern of the Y-linked PRY gene suggests a function in apoptosis but not in spermatogenesis. Mol Hum Reprod 2004, 10, (1), 15-21. 58. Reijo, R.; Lee, T. Y.; Salo, P.; Alagappan, R.; Brown, L. G.; Rosenberg, M.; Rozen, S.; Jaffe, T.; Straus, D.; Hovatta, O.; et al., Diverse spermatogenic defects in humans caused by Y chromosome deletions encompassing a novel RNA-binding protein gene. Nat Genet 1995, 10, (4), 383-93. 59. Reynolds, N.; Cooke, H. J., Role of the DAZ genes in male fertility. Reprod Biomed Online 2005, 10, (1), 72-80. 60. Kee, K.; Angeles, V. T.; Flores, M.; Nguyen, H. N.; Reijo Pera, R. A., Human DAZL, DAZ and BOULE genes modulate primordial germ-cell and haploid gamete formation. Nature 2009, 462, (7270), 222-5. 61. Slee, R.; Grimes, B.; Speed, R. M.; Taggart, M.; Maguire, S. M.; Ross, A.; McGill, N. I.; Saunders, P. T.; Cooke, H. J., A human DAZ transgene confers partial rescue of the mouse Dazl null phenotype. Proc Natl Acad Sci U S A 1999, 96, (14), 8040-5. 62. Lahn, B. T.; Tang, Z. L.; Zhou, J.; Barndt, R. J.; Parvinen, M.; Allis, C. D.; Page, D. C., Previously uncharacterized histone acetyltransferases implicated in mammalian spermatogenesis. Proc Natl Acad Sci U S A 2002, 99, (13), 8707-12. 63. Tse, J. Y.; Wong, E. Y.; Cheung, A. N.; O, W. S.; Tam, P. C.; Yeung, W. S., Specific expression of VCY2 in human male germ cells and its involvement in the pathogenesis of male infertility. Biol Reprod 2003, 69, (3), 746-51. 64. Wong, E. Y.; Tse, J. Y.; Yao, K. M.; Tam, P. C.; Yeung, W. S., VCY2 protein interacts with the HECT domain of ubiquitin-protein ligase E3A. Biochem Biophys Res Commun 2002, 296, (5), 1104-11. 65. Lardone, M. C.; Parodi, D. A.; Valdevenito, R.; Ebensperger, M.; Piottante, A.; Madariaga, M.; Smith, R.; Pommer, R.; Zambrano, N.; Castro, A., Quantification of 30

ACS Paragon Plus Environment

Page 31 of 44

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

DDX3Y, RBMY1, DAZ and TSPY mRNAs in testes of patients with severe impairment of spermatogenesis. Mol Hum Reprod 2007, 13, (10), 705-12. 66. Grgurevic, N.; Majdic, G., Sex differences in the brain-an interplay of sex steroid hormones and sex chromosomes. Clin Sci (Lond) 2016, 130, (17), 1481-97. 67. Johansson, M. M.; Lundin, E.; Qian, X.; Mirzazadeh, M.; Halvardson, J.; Darj, E.; Feuk, L.; Nilsson, M.; Jazin, E., Spatial sexual dimorphism of X and Y homolog gene expression in the human central nervous system during early male development. Biol Sex Differ 2016, 7, 5. 68. Loke, H.; Harley, V.; Lee, J., Biological factors underlying sex differences in neurological disorders. Int J Biochem Cell Biol 2015, 65, 139-50. 69. Dennis, C., Brain development: the most important sexual organ. Nature 2004, 427, (6973), 390-2. 70. Dewing, P.; Chiang, C. W.; Sinchak, K.; Sim, H.; Fernagut, P. O.; Kelly, S.; Chesselet, M. F.; Micevych, P. E.; Albrecht, K. H.; Harley, V. R.; Vilain, E., Direct regulation of adult brain function by the male-specific factor SRY. Curr Biol 2006, 16, (4), 415-20. 71. Sekido, R., The potential role of SRY in epigenetic gene regulation during brain sexual differentiation in mammals. Adv Genet 2014, 86, 135-65. 72. Weickert, C. S.; Elashoff, M.; Richards, A. B.; Sinclair, D.; Bahn, S.; Paabo, S.; Khaitovich, P.; Webster, M. J., Transcriptome analysis of male-female differences in prefrontal cortical development. Mol Psychiatry 2009, 14, (6), 558-61. 73. Kang, H. J.; Kawasawa, Y. I.; Cheng, F.; Zhu, Y.; Xu, X.; Li, M.; Sousa, A. M.; Pletikos, M.; Meyer, K. A.; Sedmak, G.; Guennel, T.; Shin, Y.; Johnson, M. B.; Krsnik, Z.; Mayer, S.; Fertuzinhos, S.; Umlauf, S.; Lisgo, S. N.; Vortmeyer, A.; Weinberger, D. R.; Mane, S.; Hyde, T. M.; Huttner, A.; Reimers, M.; Kleinman, J. E.; Sestan, N., Spatiotemporal transcriptome of the human brain. Nature 2011, 478, (7370), 483-9. 74. Czech, D. P.; Lee, J.; Sim, H.; Parish, C. L.; Vilain, E.; Harley, V. R., The human testis-determining factor SRY localizes in midbrain dopamine neurons and regulates multiple components of catecholamine synthesis and metabolism. J Neurochem 2012, 122, (2), 260-71. 75. Priddle, T. H.; Crow, T. J., Protocadherin 11X/Y a human-specific gene pair: an immunohistochemical survey of fetal and adult brains. Cereb Cortex 2013, 23, (8), 1933-41. 76. Priddle, T. H.; Crow, T. J., The protocadherin 11X/Y (PCDH11X/Y) gene pair as determinant of cerebral asymmetry in modern Homo sapiens. Ann N Y Acad Sci 2013, 1288, 36-47. 77. Blanco, P.; Sargent, C. A.; Boucher, C. A.; Mitchell, M.; Affara, N. A., Conservation of PCDHX in mammals; expression of human X/Y genes predominantly in brain. Mamm Genome 2000, 11, (10), 906-14. 78. Vanhalst, K.; Kools, P.; Staes, K.; van Roy, F.; Redies, C., delta-Protocadherins: a gene family expressed differentially in the mouse brain. Cell Mol Life Sci 2005, 62, (11), 1247-59. 79. Beddington, R. S.; Robertson, E. J., Axis development and early asymmetry in mammals. Cell 1999, 96, (2), 195-209. 80. Ding, Y.; Ploper, D.; Sosa, E. A.; Colozza, G.; Moriyama, Y.; Benitez, M. D.; Zhang, K.; Merkurjev, D.; De Robertis, E. M., Spemann organizer transcriptome 31

ACS Paragon Plus Environment

Journal of Proteome Research

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 32 of 44

induction by early beta-catenin, Wnt, Nodal, and Siamois signals in Xenopus laevis. Proc Natl Acad Sci U S A 2017, 114, (15), E3081-E3090. 81. Husken, U.; Carl, M., The Wnt/beta-catenin signaling pathway establishes neuroanatomical asymmetries and their laterality. Mech Dev 2013, 130, (6-8), 330-5. 82. Petersen, C. P.; Reddien, P. W., Wnt signaling and the polarity of the primary body axis. Cell 2009, 139, (6), 1056-68. 83. Bottos, A.; Rissone, A.; Bussolino, F.; Arese, M., Neurexins and neuroligins: synapses look out of the nervous system. Cell Mol Life Sci 2011, 68, (16), 2655-66. 84. Hoon, M.; Soykan, T.; Falkenburger, B.; Hammer, M.; Patrizi, A.; Schmidt, K. F.; Sassoe-Pognetto, M.; Lowel, S.; Moser, T.; Taschenberger, H.; Brose, N.; Varoqueaux, F., Neuroligin-4 is localized to glycinergic postsynapses and regulates inhibition in the retina. Proc Natl Acad Sci U S A 2011, 108, (7), 3053-8. 85. Ross, J. L.; Tartaglia, N.; Merry, D. E.; Dalva, M.; Zinn, A. R., Behavioral phenotypes in males with XYY and possible role of increased NLGN4Y expression in autism features. Genes Brain Behav 2015, 14, (2), 137-44. 86. Bishop, D. V.; Jacobs, P. A.; Lachlan, K.; Wellesley, D.; Barnicoat, A.; Boyd, P. A.; Fryer, A.; Middlemiss, P.; Smithson, S.; Metcalfe, K.; Shears, D.; Leggett, V.; Nation, K.; Scerif, G., Autism, language and communication in children with sex chromosome trisomies. Arch Dis Child 2011, 96, (10), 954-9. 87. Zhang, C.; Milunsky, J. M.; Newton, S.; Ko, J.; Zhao, G.; Maher, T. A.; TagerFlusberg, H.; Bolliger, M. F.; Carter, A. S.; Boucard, A. A.; Powell, C. M.; Sudhof, T. C., A neuroligin-4 missense mutation associated with autism impairs neuroligin-4 folding and endoplasmic reticulum export. J Neurosci 2009, 29, (35), 10843-54. 88. Blenck, C. L.; Harvey, P. A.; Reckelhoff, J. F.; Leinwand, L. A., The Importance of Biological Sex and Estrogen in Rodent Models of Cardiovascular Health and Disease. Circ Res 2016, 118, (8), 1294-312. 89. Salton, C. J.; Chuang, M. L.; O'Donnell, C. J.; Kupka, M. J.; Larson, M. G.; Kissinger, K. V.; Edelman, R. R.; Levy, D.; Manning, W. J., Gender differences and normal left ventricular anatomy in an adult population free of hypertension. A cardiovascular magnetic resonance study of the Framingham Heart Study Offspring cohort. J Am Coll Cardiol 2002, 39, (6), 1055-60. 90. Isensee, J.; Witt, H.; Pregla, R.; Hetzer, R.; Regitz-Zagrosek, V.; Noppinger, P. R., Sexually dimorphic gene expression in the heart of mice and men. J Mol Med (Berl) 2008, 86, (1), 61-74. 91. Vijay, V.; Han, T.; Moland, C. L.; Kwekel, J. C.; Fuscoe, J. C.; Desai, V. G., Sexual dimorphism in the expression of mitochondria-related genes in rat heart at different ages. PLoS One 2015, 10, (1), e0117047. 92. Roger, V. L.; Go, A. S.; Lloyd-Jones, D. M.; Benjamin, E. J.; Berry, J. D.; Borden, W. B.; Bravata, D. M.; Dai, S.; Ford, E. S.; Fox, C. S.; Fullerton, H. J.; Gillespie, C.; Hailpern, S. M.; Heit, J. A.; Howard, V. J.; Kissela, B. M.; Kittner, S. J.; Lackland, D. T.; Lichtman, J. H.; Lisabeth, L. D.; Makuc, D. M.; Marcus, G. M.; Marelli, A.; Matchar, D. B.; Moy, C. S.; Mozaffarian, D.; Mussolino, M. E.; Nichol, G.; Paynter, N. P.; Soliman, E. Z.; Sorlie, P. D.; Sotoodehnia, N.; Turan, T. N.; Virani, S. S.; Wong, N. D.; Woo, D.; Turner, M. B.; American Heart Association Statistics, C.; Stroke Statistics, S., Executive summary: heart disease and stroke statistics--2012 update: a report from the American Heart Association. Circulation 2012, 125, (1), 188-97. 32

ACS Paragon Plus Environment

Page 33 of 44

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

93. Fairweather, D.; Cooper, L. T., Jr.; Blauwet, L. A., Sex and gender differences in myocarditis and dilated cardiomyopathy. Curr Probl Cardiol 2013, 38, (1), 7-46. 94. Cooper, L. T.; Mather, P. J.; Alexis, J. D.; Pauly, D. F.; Torre-Amione, G.; Wittstein, I. S.; Dec, G. W.; Zucker, M.; Narula, J.; Kip, K.; McNamara, D. M.; Investigators, I., Myocardial recovery in peripartum cardiomyopathy: prospective comparison with recent onset cardiomyopathy in men and nonperipartum women. J Card Fail 2012, 18, (1), 28-33. 95. Heidecker, B.; Lamirault, G.; Kasper, E. K.; Wittstein, I. S.; Champion, H. C.; Breton, E.; Russell, S. D.; Hall, J.; Kittleson, M. M.; Baughman, K. L.; Hare, J. M., The gene expression profile of patients with new-onset heart failure reveals important gender-specific differences. Eur Heart J 2010, 31, (10), 1188-96. 96. Babiker, F. A.; De Windt, L. J.; van Eickels, M.; Grohe, C.; Meyer, R.; Doevendans, P. A., Estrogenic hormone action in the heart: regulatory network and function. Cardiovasc Res 2002, 53, (3), 709-19. 97. Du, X. J., Gender modulates cardiac phenotype development in genetically modified mice. Cardiovasc Res 2004, 63, (3), 510-9. 98. Praktiknjo, S. D.; Picard, S.; Deschepper, C. F., Comparisons of chromosome Ysubstituted mouse strains reveal that the male-specific chromosome modulates the effects of androgens on cardiac functions. Biol Sex Differ 2016, 7, 61. 99. Arnold, A. P., Conceptual frameworks and mouse models for studying sex differences in physiology and disease: why compensation changes the game. Exp Neurol 2014, 259, 2-9. 100. Llamas, B.; Belanger, S.; Picard, S.; Deschepper, C. F., Cardiac mass and cardiomyocyte size are governed by different genetic loci on either autosomes or chromosome Y in recombinant inbred mice. Physiol Genomics 2007, 31, (2), 176-82. 101. Nadeau, J. H.; Burrage, L. C.; Restivo, J.; Pao, Y. H.; Churchill, G.; Hoit, B. D., Pleiotropy, homeostasis, and functional networks based on assays of cardiovascular traits in genetically randomized populations. Genome Res 2003, 13, (9), 2082-91. 102. Llamas, B.; Verdugo, R. A.; Churchill, G. A.; Deschepper, C. F., Chromosome Y variants from different inbred mouse strains are linked to differences in the morphologic and molecular responses of cardiac cells to postpubertal testosterone. BMC Genomics 2009, 10, 150. 103. Praktiknjo, S. D.; Llamas, B.; Scott-Boyer, M. P.; Picard, S.; Robert, F.; Langlais, D.; Haibe-Kains, B.; Faubert, D.; Silversides, D. W.; Deschepper, C. F., Novel effects of chromosome Y on cardiac regulation, chromatin remodeling, and neonatal programming in male mice. Endocrinology 2013, 154, (12), 4746-56. 104. Lee, S.; Lee, J. W.; Lee, S. K., UTX, a histone H3-lysine 27 demethylase, acts as a critical switch to activate the cardiac developmental program. Dev Cell 2012, 22, (1), 25-37. 105. Wang, C.; Lee, J. E.; Cho, Y. W.; Xiao, Y.; Jin, Q.; Liu, C.; Ge, K., UTX regulates mesoderm differentiation of embryonic stem cells independent of H3K27 demethylase activity. Proc Natl Acad Sci U S A 2012, 109, (38), 15324-9. 106. Morales Torres, C.; Laugesen, A.; Helin, K., Utx is required for proper induction of ectoderm and mesoderm during differentiation of embryonic stem cells. PLoS One 2013, 8, (4), e60020. 33

ACS Paragon Plus Environment

Journal of Proteome Research

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 34 of 44

107. Dal-Pra, S.; Hodgkinson, C. P.; Mirotsou, M.; Kirste, I.; Dzau, V. J., Demethylation of H3K27 Is Essential for the Induction of Direct Cardiac Reprogramming by miR Combo. Circ Res 2017, 120, (9), 1403-1413. 108. Perissi, V.; Scafoglio, C.; Zhang, J.; Ohgi, K. A.; Rose, D. W.; Glass, C. K.; Rosenfeld, M. G., TBL1 and TBLR1 phosphorylation on regulated gene promoters overcomes dual CtBP and NCoR/SMRT transcriptional repression checkpoints. Mol Cell 2008, 29, (6), 755-66. 109. Tagariello, A.; Breuer, C.; Birkner, Y.; Schmidt, S.; Koch, A. M.; Cesnjevar, R.; Ruffer, A.; Dittrich, S.; Schneider, H.; Winterpacht, A.; Sticht, H.; Dotsch, J.; Toka, O., Functional null mutations in the gonosomal homologue gene TBL1Y are associated with non-syndromic coarctation of the aorta. Curr Mol Med 2012, 12, (2), 199-205. 110. Hilton, E.; Johnston, J.; Whalen, S.; Okamoto, N.; Hatsukawa, Y.; Nishio, J.; Kohara, H.; Hirano, Y.; Mizuno, S.; Torii, C.; Kosaki, K.; Manouvrier, S.; Boute, O.; Perveen, R.; Law, C.; Moore, A.; Fitzpatrick, D.; Lemke, J.; Fellmann, F.; Debray, F. G.; Dastot-Le-Moal, F.; Gerard, M.; Martin, J.; Bitoun, P.; Goossens, M.; Verloes, A.; Schinzel, A.; Bartholdi, D.; Bardakjian, T.; Hay, B.; Jenny, K.; Johnston, K.; Lyons, M.; Belmont, J. W.; Biesecker, L. G.; Giurgea, I.; Black, G., BCOR analysis in patients with OFCD and Lenz microphthalmia syndromes, mental retardation with ocular anomalies, and cardiac laterality defects. Eur J Hum Genet 2009, 17, (10), 1325-35. 111. Zhu, X.; Dai, F. R.; Wang, J.; Zhang, Y.; Tan, Z. P.; Zhang, Y., Novel BCOR mutation in a boy with Lenz microphthalmia/oculo-facio-cardio-dental (OFCD) syndrome. Gene 2015, 571, (1), 142-4. 112. Sabolic, I.; Asif, A. R.; Budach, W. E.; Wanke, C.; Bahn, A.; Burckhardt, G., Gender differences in kidney function. Pflugers Arch 2007, 455, (3), 397-429. 113. Munger, K.; Baylis, C., Sex differences in renal hemodynamics in rats. Am J Physiol 1988, 254, (2 Pt 2), F223-31. 114. Neugarten, J.; Acharya, A.; Silbiger, S. R., Effect of gender on the progression of nondiabetic renal disease: a meta-analysis. J Am Soc Nephrol 2000, 11, (2), 319-29. 115. Elliot, S. J.; Berho, M.; Korach, K.; Doublier, S.; Lupia, E.; Striker, G. E.; Karl, M., Gender-specific effects of endogenous testosterone: female alpha-estrogen receptordeficient C57Bl/6J mice develop glomerulosclerosis. Kidney Int 2007, 72, (4), 464-72. 116. Xu, J.; Burgoyne, P. S.; Arnold, A. P., Sex differences in sex chromosome gene expression in mouse brain. Hum Mol Genet 2002, 11, (12), 1409-19. 117. Si, H.; Banga, R. S.; Kapitsinou, P.; Ramaiah, M.; Lawrence, J.; Kambhampati, G.; Gruenwald, A.; Bottinger, E.; Glicklich, D.; Tellis, V.; Greenstein, S.; Thomas, D. B.; Pullman, J.; Fazzari, M.; Susztak, K., Human and murine kidneys show gender- and species-specific gene expression differences in response to injury. PLoS One 2009, 4, (3), e4802. 118. Hilliard, L. M.; Sampson, A. K.; Brown, R. D.; Denton, K. M., The "his and hers" of the renin-angiotensin system. Curr Hypertens Rep 2013, 15, (1), 71-9. 119. Reckelhoff, J. F., Gender differences in the regulation of blood pressure. Hypertension 2001, 37, (5), 1199-208. 120. Ely, D. L.; Turner, M. E., Hypertension in the spontaneously hypertensive rat is linked to the Y chromosome. Hypertension 1990, 16, (3), 277-81. 121. Turner, M. E.; Farkas, J.; Dunmire, J.; Ely, D.; Milsted, A., Which Sry locus is the hypertensive Y chromosome locus? Hypertension 2009, 53, (2), 430-5. 34

ACS Paragon Plus Environment

Page 35 of 44

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

122. Milsted, A.; Underwood, A. C.; Dunmire, J.; DelPuerto, H. L.; Martins, A. S.; Ely, D. L.; Turner, M. E., Regulation of multiple renin-angiotensin system genes by Sry. J Hypertens 2010, 28, (1), 59-64. 123. Prokop, J. W.; Rauscher, F. J., 3rd; Peng, H.; Liu, Y.; Araujo, F. C.; Watanabe, I.; Reis, F. M.; Milsted, A., MAS promoter regulation: a role for Sry and tyrosine nitration of the KRAB domain of ZNF274 as a feedback mechanism. Clin Sci (Lond) 2014, 126, (10), 727-38. 124. Araujo, F. C.; Milsted, A.; Watanabe, I. K.; Del Puerto, H. L.; Santos, R. A.; Lazar, J.; Reis, F. M.; Prokop, J. W., Similarities and differences of X and Y chromosome homologous genes, SRY and SOX3, in regulating the renin-angiotensin system promoters. Physiol Genomics 2015, 47, (5), 177-86. 125. Yanai, K.; Saito, T.; Kakinuma, Y.; Kon, Y.; Hirota, K.; Taniguchi-Yanai, K.; Nishijo, N.; Shigematsu, Y.; Horiguchi, H.; Kasuya, Y.; Sugiyama, F.; Yagami, K.; Murakami, K.; Fukamizu, A., Renin-dependent cardiovascular functions and reninindependent blood-brain barrier functions revealed by renin-deficient mice. J Biol Chem 2000, 275, (1), 5-8. 126. Niimura, F.; Labosky, P. A.; Kakuchi, J.; Okubo, S.; Yoshida, H.; Oikawa, T.; Ichiki, T.; Naftilan, A. J.; Fogo, A.; Inagami, T.; et al., Gene targeting in mice reveals a requirement for angiotensin in the development and maintenance of kidney morphology and growth factor regulation. J Clin Invest 1995, 96, (6), 2947-54. 127. Chen, Y.; Lasaitiene, D.; Friberg, P., The renin-angiotensin system in kidney development. Acta Physiol Scand 2004, 181, (4), 529-35. 128. Li, X.; Li, H. P.; Amsler, K.; Hyink, D.; Wilson, P. D.; Burrow, C. R., PRKX, a phylogenetically and functionally distinct cAMP-dependent protein kinase, activates renal epithelial cell migration and morphogenesis. Proc Natl Acad Sci U S A 2002, 99, (14), 9260-5. 129. Li, X.; Hyink, D. P.; Radbill, B.; Sudol, M.; Zhang, H.; Zheleznova, N. N.; Wilson, P. D., Protein kinase-X interacts with Pin-1 and Polycystin-1 during mouse kidney development. Kidney Int 2009, 76, (1), 54-62. 130. Huang, S.; Li, Q.; Alberts, I.; Li, X., PRKX, a Novel cAMP-Dependent Protein Kinase Member, Plays an Important Role in Development. J Cell Biochem 2016, 117, (3), 566-73. 131. Li, X.; Burrow, C. R.; Polgar, K.; Hyink, D. P.; Gusella, G. L.; Wilson, P. D., Protein kinase X (PRKX) can rescue the effects of polycystic kidney disease-1 gene (PKD1) deficiency. Biochim Biophys Acta 2008, 1782, (1), 1-9. 132. Omenn, G. S.; Lane, L.; Lundberg, E. K.; Beavis, R. C.; Nesvizhskii, A. I.; Deutsch, E. W., Metrics for the Human Proteome Project 2015: progress on the human proteome and guidelines for high-confidence protein identification. Journal of proteome research 2015, 14, (9), 3452-3460.

35

ACS Paragon Plus Environment

Journal of Proteome Research

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 36 of 44

Figure legends

Figure 1. Y chromosome Genes and pathways required for the testis development and spermatogenesis. Bipotential gonadal ridge develops into one of two functionally distinct organs, a testis or an ovary. Genes such as Zfpm2, Wt1, Nr5a1, Cbx2, Jmjd1a and Dax1 are essential for the correct initiation and upregulation of SRY. SRY as a master gene initiates a cascade of events preceding testis differentiation and ovarian developmental inhibition by repressing Wnt signaling-dependent gene expression. In male genital ridge, SRY expression at its critical level is accompanied by sexually dimorphic expression of its downstream target, SOX9. In addition to SRY, Nr5a1 and Dax1 regulate SOX9 expression. Sex-determining genes activate the downstream targets such as Amh, Vnn, Ptgts, Cbln4, Fgf9, Gdnf, Cyp26b1, Wnt5a, Dhh and Ntf3 that are necessary for the regulation and maintenance of this crucial testis formation pathway. Human Y genes play an important role in spermatogenesis and their deletions result in spermatogenic defects. DDX3Y and TSPY is expressed in spermatogonia, RBMY, DAZ and BPY2 in spermatogonia, spermatocyte and round spermatid, USP9Y and CDY in spermatids, and PRY in spermatids and spermatozoa.

Figure 2. Heatmap presentation of the transcript profile of MSY genes during human brain development. All MSY gene expression data in male brain regions was downloaded from BrainSpan (atlas of developing human brain) covering prenatal (8-38 pcw), infancy (birth-18 mos), childhood (19 mos-11 yrs), adolescence (12-19 yrs) and adulthood (20-60+ yrs) developmental stages. The expression level of genes is shown 36

ACS Paragon Plus Environment

Page 37 of 44

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

in log2 RPKM (reads per kilobase per million) value. Abbreviations; pcw (post conception weeks), mos (months), yrs (years), DFC (dorsolateral prefrontal cortex), VFC (ventrolateral prefrontal cortex), MFC (medial prefrontal cortex), OFC (orbital frontal cortex), M1C (primary motor cortex), S1C (primary somatosensory cortex), IPC (posterior inferior parietal cortex), A1C (primary auditory cortex), STC (posterior(caudal) superior temporal cortex), ITC (inferolateral temporal cortex), V1C (primary visual cortex), HIP (hippocampus), AMY (amygdaloid complex), STR (striatum), MD (mediodorsal nucleus of thalamus) and CBC (cerebellar cortex), P (prenatal), I (infancy), C (childhood), T (adolescence or teenage years) and A (adulthood).

Figure 3. Visual representation of dysregulated MSY transcripts during cardiac differentiation of human embryonic stem cell (hESC). The Circos map compares the MSY gene expression pattern in hESC, mesendoderm, cardiac mesoderm, cardiac progenitor cell and cardiomyocyte. The inner colored segments below the outer segments are representative for each specific gene or differentiation stages; for instance, the red segment is related to the BCORP1 transcript, and the green segment corresponds to the mesendoderm stage. The outer segment of each gene demonstrates in which differentiation stages it’s mainly expressed. For example, BCORP1 mainly expressed in cardiac mesoderm stage (30%), cardiac progenitor stage (26%), cardiomyocytes (25%), respectively. The outer segment of each differentiation stage presents the contribution level of MSY genes in the expression of genes in each group (i.e. in cardiac mesoderm stage, among MSY genes, BCORP1 has the highest fold change ~ 11 %). The inner arcs link the expression of the genes to differentiation 37

ACS Paragon Plus Environment

Journal of Proteome Research

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 38 of 44

stages. The inner segment of genes sorted in a descending order for instance BCORP1 showed the maximal overexpression (0-50) specifically at cardiac mesoderm (~ 15%) while SRY showed the maximal downregulation (0-10) during cardiac differentiation. The inner segment of developmental stages show that most of Y chromosome genes overexpress at cardiac mesoderm stage (0-150) followed by other stages. Gene expression data obtained from REF.24.

Figure 4.

TBL1Y may play role in cardiac development by Notch signaling

activation. Cell to cell contacts and subsequent interaction of NOTCH receptor and its ligand, JAGGED 1, result in Notch signaling activation. Activation of this signaling requires the release of a dual repression caused by distinct corepressor complexes, CtBP and NCoR/SMRT/HDAC. TBL1X/TBL1Y and TBLR1 are F box/WD40 containing factors that function as exchangers in the release of dual repression by dismissal of CtBP and NCoR/SMART from corepressor complex resulting in transcriptional activation of Notch target genes such as Hes1. MAML stabilizes the RBP-J/NICD binding to DNA during activation of target genes.

38

ACS Paragon Plus Environment

Page 39 of 44

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

Table 1. Y chromosome genes, their status in neXtProt and findings on their roles in development 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48

Y-Chr genes USP9Y VCY BPY2 EIF1AY PRY TMSB4Y UTY DDX3Y ZFY RBMY1A1 RBMY1C RPS4Y1 CD24 TSPY1 SRY DAZ2 RBMY1F DAZ4 TGIF2LY HSFY KDM5D PCDH11Y DAZ1 DAZ3 CDY2 CDY1 XKRY2 RBMY1B RBMY1E XKRY RBMY1D NLGN4Y RPS4Y2 AMELY TBL1Y PRORY TSPY2 TSPY3 TSPY4 TSPY8 TSPY10 SLC9B1P1 PRKY BCORP1 TTTY13 TTTY12 TTTY10 TXNLGY

PE level PE1 PE1 PE1 PE1 PE1 PE1 PE1 PE1 PE1 PE1 PE1 PE1 PE1 PE1 PE1 PE1 PE1 PE1 PE1 PE1 PE1 PE1 PE1 PE1 PE1 PE1 PE2 PE2 PE2 PE2 PE2 PE2 PE2 PE2 PE2 PE2 PE3 PE3 PE3 PE3 PE3 PE5 PE5 PE5 PE5 PE5 PE5 PE5

Function ubiquitin-specific protease, Hydrolase unknown function Protein binding Translational initiation, Protein binding unknown function Actin monomer binding histone demethylase RNA helicase, DNA binding, RNA binding transcriptional activator RNA splicing, Protein binding RNA binding, RNA splicing RNA binding, rRNA binding Signal transducer activity, Protein binding, Protein binding Transcription factor, DNA binding RNA binding RNA binding, RNA splicing RNA binding DNA binding, protein binding Transcription factor, , protein binding Histone demethylase, DNA binding Calcium ion binding RNA binding, translation activator RNA binding Histone acetyltransferase Histone acetyltransferase unknown function RNA binding, mRNA splicing RNA binding, mRNA splicing unknown function RNA binding, mRNA splicing Cell adhesion molecule binding Translation. rRNA binding Structural constituent of tooth enamel Transcription activation/corepression unknown function unknown function unknown function unknown function unknown function unknown function proton antiporter activity Protein serine/threonine kinase unknown function unknown function unknown function unknown function Syntaxin binding

39

ACS Paragon Plus Environment

Human organogenesis Brain, spermatogenesis, heart spermatogenesis Brain spermatogenesis, heart Brain Brain, heart Brain, spermatogenesis, heart Brain, heart spermatogenesis, heart spermatogenesis, heart Brain, heart Brain spermatogenesis Brain, testis, kidney spermatogenesis spermatogenesis, heart spermatogenesis Brain, heart Brain, heart Brain, heart spermatogenesis spermatogenesis spermatogenesis spermatogenesis spermatogenesis, heart spermatogenesis, heart Single fertilization spermatogenesis, heart Brain Brain Tooth enamel development Brain, heart spermatogenesis spermatogenesis spermatogenesis spermatogenesis spermatogenesis Brain, kidney heart Brain Brain

Journal of Proteome Research

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Figure 1. Y chromosome Genes and pathways required for the testis development and spermatogenesis. Bipotential gonadal ridge develops into one of two functionally distinct organs, a testis or an ovary. Genes such as Zfpm2, Wt1, Nr5a1, Cbx2, Jmjd1a and Dax1 are essential for the correct initiation and upregulation of SRY. SRY as a master gene initiates a cascade of events preceding testis differentiation and ovarian developmental inhibition by repressing Wnt signaling-dependent gene expression. In male genital ridge, SRY expression at its critical level is accompanied by sexually dimorphic expression of its downstream target, SOX9. In addition to SRY, Nr5a1 and Dax1 regulate SOX9 expression. Sex-determining genes activate the downstream targets such as Amh, Vnn, Ptgts, Cbln4, Fgf9, Gdnf, Cyp26b1, Wnt5a, Dhh and Ntf3 that are necessary for the regulation and maintenance of this crucial testis formation pathway. Human Y genes play an important role in spermatogenesis and their deletions result in spermatogenic defects. DDX3Y and TSPY is expressed in spermatogonia, RBMY, DAZ and BPY2 in spermatogonia, spermatocyte and round spermatid, USP9Y and CDY in spermatids, and PRY in spermatids and spermatozoa. 361x270mm (300 x 300 DPI)

ACS Paragon Plus Environment

Page 40 of 44

Page 41 of 44

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

Figure 2. Heatmap presentation of the transcript profile of MSY genes during human brain development. All MSY gene expression data in male brain regions was downloaded from BrainSpan (atlas of developing human brain) covering prenatal (8-38 pcw), infancy (birth-18 mos), childhood (19 mos-11 yrs), adolescence (12-19 yrs) and adulthood (20-60+ yrs) developmental stages. The expression level of genes is shown in log2 RPKM (reads per kilobase per million) value. Abbreviations; pcw (post conception weeks), mos (months), yrs (years), DFC (dorsolateral prefrontal cortex), VFC (ventrolateral prefrontal cortex), MFC (medial prefrontal cortex), OFC (orbital frontal cortex), M1C (primary motor cortex), S1C (primary somatosensory cortex), IPC (posterior inferior parietal cortex), A1C (primary auditory cortex), STC (posterior(caudal) superior temporal cortex), ITC (inferolateral temporal cortex), V1C (primary visual cortex), HIP (hippocampus), AMY (amygdaloid complex), STR (striatum), MD (mediodorsal nucleus of thalamus) and CBC (cerebellar cortex), P (prenatal), I (infancy), C (childhood), T (adolescence or teenage years) and A (adulthood). 361x270mm (300 x 300 DPI)

ACS Paragon Plus Environment

Journal of Proteome Research

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Figure 3. Visual representation of dysregulated MSY transcripts during cardiac differentiation of human embryonic stem cell (hESC). The Circos map compares the MSY gene expression pattern in hESC, mesendoderm, cardiac mesoderm, cardiac progenitor cell and cardiomyocyte. The inner colored segments below the outer segments are representative for each specific gene or differentiation stages; for instance, the red segment is related to the BCORP1 transcript, and the green segment corresponds to the mesendoderm stage. The outer segment of each gene demonstrates in which differentiation stages it’s mainly expressed. For example, BCORP1 mainly expressed in cardiac mesoderm stage (30%), cardiac progenitor stage (26%), cardiomyocytes (25%), respectively. The outer segment of each differentiation stage presents the contribution level of MSY genes in the expression of genes in each group (i.e. in cardiac mesoderm stage, among MSY genes, BCORP1 has the highest fold change ~ 11 %). The inner arcs link the expression of the genes to differentiation stages. The inner segment of genes sorted in a descending order for instance BCORP1 showed the maximal overexpression (0-50) specifically at cardiac mesoderm (~ 15%) while SRY showed the maximal downregulation (0-10) during cardiac differentiation. The inner segment of developmental stages show that most of Y chromosome genes overexpress at cardiac mesoderm stage (0150) followed by other stages. Gene expression data obtained from REF.24. 199x199mm (300 x 300 DPI)

ACS Paragon Plus Environment

Page 42 of 44

Page 43 of 44

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

Figure 4. TBL1Y may play role in cardiac development by Notch signaling activation. Cell to cell contacts and subsequent interaction of NOTCH receptor and its ligand, JAGGED 1, result in Notch signaling activation. Activation of this signaling requires the release of a dual repression caused by distinct corepressor complexes, CtBP and NCoR/SMRT/HDAC. TBL1X/TBL1Y and TBLR1 are F box/WD40 containing factors that function as exchangers in the release of dual repression by dismissal of CtBP and NCoR/SMART from corepressor complex resulting in transcriptional activation of Notch target genes such as Hes1. MAML stabilizes the RBP-J/NICD binding to DNA during activation of target genes.

468x417mm (144 x 144 DPI)

ACS Paragon Plus Environment

Journal of Proteome Research

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

for TOC only 190x275mm (300 x 300 DPI)

ACS Paragon Plus Environment

Page 44 of 44