Pax6 expressed in osteocytes inhibits canonical wnt signaling · 2020. 7. 3. · mamayya, bangaaru...

57
Pax6 expressed in osteocytes inhibits canonical wnt signaling Ajita Jami Department of Medical Science The Graduate School, Yonsei University

Transcript of Pax6 expressed in osteocytes inhibits canonical wnt signaling · 2020. 7. 3. · mamayya, bangaaru...

  • Pax6 expressed in osteocytes

    inhibits canonical wnt signaling

    Ajita Jami

    Department of Medical Science

    The Graduate School, Yonsei University

  • Pax6 expressed in osteocytes

    inhibits canonical wnt signaling

    Directed by Professor Sung Kil Lim

    The Doctoral Dissertation submitted to

    the Department of Medical Science,

    the Graduate school of Yonsei University

    in partial fulfillment of the requirements for

    the degree of Doctor of Philosophy

    Ajita Jami

    June 2013

  • ACKNOWLEDGEMENTS

    “GURU BRHAMA

    GURUUR VISHUNUHU

    GURUR DEVO MAHESWARA”

    - VEDA

    I begin my acknowledgement by thanking almighty Teachers whose

    blessing has enabled me to fulfill my thesis work with bundle of joy.

    It is my proud privilege to express my deep sense of gratitude to my

    respected and esteemed teacher and supervisor, Professor Sung Kil Lim for his

    invaluable contributions, guidance and cooperation held up at each and every stage

    of this work. I am extremely indebted and grateful to him for his realistic approach,

    honest advice and sincere heartfelt motivation of guiding and taking interest in this

    work. He guided me right from the protocol preparation and presentation up to the

    completion of this research work, ready to solve each and every problem that might

    have come on my way while pursuing the work. In spite of his busy and hectic

    schedule, he was kind enough to be available for the correction of my thesis work.

    Last but no the least, it was with his blessings and I could accomplish my work.

    I extend my heartfelt thanks to the committee members Prof. Myoung Hee

    Kim, Prof. Han Sung Jung, Prof. Sahng wook park, Prof. Jin Woo Lee for their

    valuable suggestions and advices for my thesis work.

    I express my deep gratitude for the admirable contribution by Dr.Gadi

    Jogeswar, who have provided extremely valuable assistance in many aspects of the

    study to made the task so very easy and simple.

    I would like to say my sincere thanks to the Brain korea 21(BK21) for

    providing me the scholarship.

    I am very much thankful to the funding sources which made successful

  • completion of my P.hD work. This work was supported by a National Research

    foundation of Korea (NRF) grant funded by the Korean government (MEST) (No.

    20120000486).

    I would like to say my heartful thanks to my lab senior, assistant research

    professor and very good friend Dr.Eun Jin Kim, who given lot of moral support,

    encouragement, valuable suggestions, guidance regarding many aspects of my work.

    Many thanks to my sister Mi Jeong Lee for her valuable suggestions, help and to

    write my thesis abstract in Korean language. I am very much thankful to Myoung

    Jin Lee sister for her friendly support. My heartful thanks to previous members of

    our lab, Dr. Sujin Park, Dr.Han Seok Choi, Dr.Kyoung Min Kim, Dr.Kwang Joon

    Kim, leelan for their help and support in this lab.

    My sincere thanks to Min Jung Lee (Dental College) for her good support

    in doing animal experiments.

    My special thanks to my brother ankamreddy harinarayana for his

    admirable help to me. I am very much thankful to my friend Dr.Kiran and his wife

    Dr.Prasanna and Dr.Mahesh for their support, encouragement and suggestions. I

    wish to thank all the professors, other members and technical support people. I am

    very much thankful to my Korean friends, Dr. Hyo Jin Kang, Moon Min Jeong, Lim

    Young Sown and others for their help during my research work.

    I am very much thankful to my previous house owners in korea Kim

    Kyoung moom uncle and Lee Su Jin aunty for their parental care and for providing

    accommodation. My sincere thanks to their children jun and jai for their help. My

    sincere thanks to my Chinese friend and sister Jin Hanglian for her help and moral

    support and suggestions.

    I thank my parents and parent-in-laws, my pedanannagaru, peddamma, my

    beloved husband (Dr. Gumma Vidyanidhi) , my son (Sourya Nanda Gopal),

    Rajuncle, Janaki aunty as they have been the main source of my energy, support and

  • encouragement. This work would not have reached its present shape without their

    constant inspiration and wishes. I would like to say my heartful thanks to my

    brothers, brother-in-laws, sisters, sister-in laws, Aanjaneyulu uncle, Laxmi-aunty,

    and all close relatives and family members (especially my grand father, ramana

    mamayya, bangaaru mamayya, desanna mamayya, ramu chinnaanna, late rambaabu

    mamayyagaru, Rajak uncle, Appala raju uncle, Demudu baabu chinnaanna,) who

    given lot of care, support, encouragement towards me from my childhood. I am

    very much thankful to my friends Dr.Ruthala kalyani, Dr.Murali Krishna,

    Dr.Kumara swamy and his wife Dr.P.V.Suneetha, Satish, Vinod kumar badireddy,

    Dr.Prachi anand kumar, Dr.P.Sunil Kumar for their moral support and

    encouragement.

    My heartful thanks to Professor V.K.Sharma, great teacher and philosopher,

    who given lot of encouragement, moral support and valuable suggestions and

    guidance to me and to my husband.

    My heartful thanks to Late Sri Sri. A. Subba Rao, Late Sri Sri Badireddy

    Maakunayudu garu, Great teachers, Philosophers. They always inspires me and tries

    to build up good state of mind and strongly insist me to help the people.

    I dedicate my thesis to my beloved family and to great teachers

    Professor V.K.Sharma and Late Sri.A.Subbarao garu

  • TABLE OF CONTENTS

    ABSTRACT 1

    I. INTRODUCTION 3

    II. MATERIALS AND METHODS 6

    1. Animal study 6

    2. Cell culture 6

    3. Osteoblast differentiation 7

    4. Alkaline Phosphatase (ALP) staining 7

    5. RT-PCR analysis 7

    6. Plasmid constructs and transfection 9

    7. Sost promoter plasmid construction 9

    8. Transfection of cells with small interfering RNA 10

    9. Cell proliferation and cell number assay 11

    10. Western blot analysis 11

    11. Preparation of Wnt-3a conditioned medium 12

    12. Chromatin immunoprecipitaion (Chip) assay 13

    13. Luciferase reporter assay for monitoring Sost promoter activity

    and Wnt signaling 14

    14. Induction of Pax6 expression by Wnt3a treatment 15

    15. Immunohistochemistry and HE staining 15

    16. Statistical analysis 16

  • III. RESULTS 17

    1. Endogenous expression of Pax6 in various types of bone cells

    and tissues 17

    2. Effect of Pax6 on Wnt inhibitor Sost and other osteocyte markers 21

    3. Effect of Pax6 on Sost promoter activity in MLOY4 cells 24

    4. Effect of Wnt3a on Pax6 mRNA expression in MLOY4 cells 29

    5. Effect of Pax6 on Wnt signaling in MLOY4 cells 30

    IV. DISCUSSION 33

    V. CONCLUSION 36

    REFERENCES 37

    ABSTRACT (IN KOREAN) 45

    PUBLICATION LIST 47

  • LIST OF FIGURES

    Figure 1. Endogenous expression of Pax6 in bone cells and tissues 19

    Figure 2. Immunohistochemical analysis 20

    Figure 3. The effects of Pax6 on osteocyte marker genes 22

    Figure 4. Effect of Pax6 on proliferation and cell viability 23

    Figure 5. The Sost promoter sequence analysis (2kb region) 25

    Figure 6. Chip PCR analysis in MLOY4 cells 26

    Figure 7. Schematic diagram of Sost promoter reporter construct 27

    Figure 8. The effect of Pax6 on Sost promoter activity 28

    Figure 9. Effect of Wnt-3a Pax6 mRNA expression 29

    Figure 10. Effect of Pax6 on Wnt signaling 31

    Figure 11. Schematic representation of the role of Pax6 in inhibiting

    Wnt signaling through Sost 32

    LIST OF TABLES

    Table 1. Primer sequences for RT-PCR and Real-time PCR 8

  • 1

    ABSTRACT

    Pax6 expressed in osteocytes inhibits canonical wnt signaling

    Ajita Jami

    Department of Medical Science

    The Graduate School, Yonsei University

    (Directed by Professor Sung Kil Lim)

    Pax6 is a transcription factor, which belongs to the paired box-containing gene

    family. It regulates developmental processes, especially in the eyes, central nervous

    tissues and craniofacial structures. However, the role of Pax6 in bone has never

    been studied. This study report that Pax6 is expressed at both the mRNA and

    protein level in the calvaria and long bones of adult mice as well as osteocyte-like

    MLOY4 cells and suppresses the canonical Wnt signaling pathway. Moreover, the

    expression levels of Pax6 were much higher in the calvaria than the long bones, and

    Pax6 was also expressed at E16 to E18 in both the calvaria and long bones.

  • 2

    Knockdown of Pax6 in MLOY4 cells did not affect cell proliferation or survival;

    however, the expression of Sost, a late osteocyte marker gene, was significantly

    decreased. In addition, the overexpression of Pax6 suppressed the canonical Wnt

    signaling pathway by enhancing the expression of Sost. Furthermore, also

    demonstrated that Pax6 binds to the Sost promoter and that stimulation of Sost

    transcription by Pax6 was dependent on a specific Pax6-binding sequence within

    the promoter. In conclusion, the results of the present study suggest that Pax6 is

    expressed in bone and may play an important role in osteocyte differentiation by

    controlling canonical Wnt signaling.

    Key words: Pax6, Osteocyte-like MLOY4 cells, MC3T3E1-cells, Sost, Wnt

    signaling.

  • 3

    Pax6 expressed in osteocytes inhibits canonical wnt signaling

    Ajita Jami

    Department of Medical Science

    The Graduate School, Yonsei University

    (Directed by Professor Sung Kil Lim)

    I. INTRODUCTION

    Skeleton in vertebrates formed by two tissues namely, bone and cartilage1.

    Osteocytes are terminally differentiated osteoblasts, derived from matrix producing

    osteoblasts. Osteoblasts move along the bone surface before localizing to a targeted,

    pre-determined location where they become embedded in the newly forming osteoid

    bone matrix. Once embedded, their dendritic processes can extend and retract, and

    the cell bodies are able to undulate within their lacunae2. Osteocytes play

  • 4

    multifunctional roles as mechanosensors, regulators of mineral homeostasis and

    coordinators of bone remodeling processes carried out by osteoblasts and

    osteoclasts3. Osteoblasts are differentiated into osteocytes through several stages

    which include osteoblasts residing on the bone surface, osteoblastic osteocytes or

    preosteocytes, osteoid osteocytes, and mature osteocytes embedded in a mineralized

    matrix4. However, the molecular and cellular mechanisms governing osteoblast and

    osteocyte differentiation are largely unknown.

    Sclerostin, the product of the Sost gene, was previously identified as an important

    negative regulator of bone formation in two rare bone sclerosing dysplasias,

    Sclerosteosis and Van Buchem disease5. Sclerostin is a secretory glycoprotein that

    plays a key role in the regulation of bone formation through Wnt signaling6.

    Moreover, expression and secretion of sclerostin have been reported in terminally

    differentiated osteocytes7. Bone formation was regulated by sequence of events

    starting by the commitment of osteoprogenitor cells, their differentiation into pre-

    osteoblasts and into mature osteoblasts whose function is to synthesize the bone

    matrix, which was then mineralized. Recently cellular, molecular and genetic

    analyses revealed that several transcription factors involved in the control of

    osteoblastogenesis8,9,10,11. In this study, transcription factors that were upregulated

    with Sost were analyzed using gene expression omnibus (GEO) analysis (in silico

    analysis)12

    . Pax6 was identified as one of the transcription factors that were

    upregulated with the Sost gene in GEO analysis.

    Pax6 belongs to the paired box-containing gene family, and acts as a transcription

    factor regulating developmental processes13

    . Pax6 is also known to regulate the

  • 5

    development of eyes, central nervous tissues and craniofacial structures14-17

    . Pax6,

    encoded by the Pax6 gene, contains two DNA-binding domains, the paired domain

    (PD) and the homeo-domain (HD), as well as a transactivation domain. In

    vertebrates, several isoforms of Pax6 are encoded by the Pax6 gene18-20

    . Two major

    isoforms are produced by the Pax6 gene by alternative splicing, Pax6(+5a) and

    Pax6(–5a). Pax6(+5a) differs from Pax6(–5a) by the presence of an exon 5a-

    encoded 14 amino acid insertion in its PD. These two isoforms demonstrate distinct

    DNA-binding properties to the promoters of their downstream target

    proteins13,18,19,21,22

    . Pax6(-5a) has been shown to influence both cell proliferation and

    neuronal differentiation, while Pax6(+5a) has only been shown to affect

    proliferation23-26

    . The canonical Wnt signaling pathway is involved in many

    developmental processes27

    . Moreover, Pax6 is known to play an important role in

    lens morphogenesis through the inhibition of canonical Wnt/β-catenin signaling in

    the lens surface ectoderm. Previously, Pax6 was shown to regulate the expression of

    negative regulators of the Wnt signaling pathway, such as secreted frizzeled-related

    protein1 (sfrp1) and secreted frizzeled-related protein 2 (sfrp2)28

    . Although the

    canonical Wnt signaling is known to play a role in deciding the fate of osteoblast

    lineage cells, as well as in the regulation of bone mass and homeostasis29

    , the role of

    Pax6 in bone has never been reported.

    In the present study, experimentally showed the expression of Pax6 in bone

    tissues and MLOY4 established osteocyte-like cell lines, the regulation of the Sost

    gene by Pax6, and its role in inhibiting Wnt signaling.

  • 6

    II. MATERIALS AND METHODS

    1. Animal study

    Embryos used in this study were obtained from time-mated adult imprinting

    control region (ICR) pregnant mice. Embryonic day 0 (E0) was designated as the

    day on which vaginal plugs were confirmed. Embryos at E16, E18 were used in this

    study.

    2. Cell culture

    Osteocyte-like MLOY4 cells were cultured in alpha- Minimal Essential Medium

    (α-MEM) (WelGENE Inc., Daegu, South Korea) containing 5% (v/v) fetal bovine

    serum (FBS) (WelGENE Inc., Daegu, South Korea), 5% (5%) neonatal calf serum

    (NCS) (WelGENE Inc., Daegu, South Korea) and 1% Penicillin-streptomycin

    (WelGENE Inc., Daegu, South Korea) as described previously30,31

    . Mouse primary

    calvarial cells were cultured as described previously32

    . Briefly, primary calvarial

    cells (Primary-osteoblasts) were prepared from the calvaria tissue (which contains

    primary-osteoblasts, mature osteoblasts, and osteocytes, which are engulfed in the

    mineralized matrix) of neonatal mice by serial digestions with Type I collagenase

    (Gibco BRL, Grand Island, NY, USA). In these serial digestions 3-5 digestion

    fractions were collected, which contains primary calvarial cells. During the culture

    period, cells were incubated at 370C in a humidified atmosphere of 5% CO2 and

    95% air and the medium was changed for every 2 days. MC3T3-E1 cells were

    cultured in α-MEM containing 10% (v/v) FBS and 1% penicillin/streptomycin.

  • 7

    3. Osteoblast differentiation

    To induce osteoblast differentiation, MC3T3-E1 cells were supplemented with α-

    MEM containing 50µg/mL ascorbic acid (Sigma, St. Louis, MO, USA) and 10mM

    β-glycerophosphate (Sigma, St. Louis, MO, USA). The differentiation medium was

    replaced for every two days.

    4. Alkaline phosphatase (ALP) staining

    To study the osteoblast differentiation, ALP staining was done using ALP

    staining kit (Sigma, St. Louis, MO, USA) according to the manufacturer’s protocol.

    5. Reverse transcription-PCR (RT-PCR) analysis

    For RT-PCR analysis, total RNA was extracted by using Trizol reagent

    (Invitrogen, Grand Island, NY, USA) according to the standard protocol. cDNA

    synthesis was performed by using MMLV-RT32

    (Promega, Madison, WI, USA))

    according to manufacturer’s protocol. In total, 1 µL of cDNA was used as the

    template for PCR amplification of β- Actin, Pax6, Pax6 isoform, Sost, DMP1 and

    E11 along with 10 pmole of primer and Go-Taq DNA polymerase (Promega,

    Madison, WI, USA). Amplification was carried out at 95°C for initial denaturation,

    followed by appropriate cycles at 95°C for 30 s (denaturation), 55°C (for β-Actin,

    Pax6, Pax6 isoform and E11) or 57°C (for DMP1 and Sost) for 30 s (annealing) and

    72°C for 30 s (polymerization). Pax6 isoform primers were designed in exon5a of

    Pax6. Amplified products were resolved on a 1.7% agarose gel. Quantitative real-

  • 8

    time PCR was performed to measure the amount of Pax6, Sost, E11 and DMP1 with

    the Applied Biosystems Real-time PCR system with SYBR green mix (Promega,

    Madison, WI, USA). In this study, the same primers were used to amplify Pax6,

    E11 and DMP1 for both real-time PCR and RT-PCR. Primer pairs were as follows

    (Table 1).

    Table 1. Primer sequences for RT-PCR and real-time PCR

    Gene name Forward Reverse

    RT-PCR

    Pax6 5’-AGTTCTTCGCAACCTGGCTA-3’ 5’-GAGCCTCAATCTGCTCTTGG-3’

    Pax6isoform 5’-TGCGACATTTCCCGAATT-3’ 5’-TCTGTCTCGGATTTCCCAAG-3’

    Sost 5’-CCTATGACGCCAAAGATGTG-3’ 5’-GTTGTGGAAGCGGGTGAG-3’

    E11 5’-AGTGTTGTTCTGGGTTTTGG-3’ 5’-GTTGTGGAAGCGGGTGAG-3’

    DMP1 5’-AGTGAGGAGGACAGCCTGAA-3’ 5’-TCCCTGTGGAGTTGCTCTCT-3’

    β-actin 5’-TTCAACACCCCAGCCATGT-3' 5’-TGTGGTACGACCAGAGGCATAC-3’

    Real-time PCR

    Gapdh 5’-AATGTGTCCGTCGTGGATCTG-3’ 5’-CAACCTGGTCCTCAGTGTAGC-3’

    Sost 5’-TCCTGAGAACAACCAGACCA-3’ 5’-GCAGCTGTACTCGGACACATC-3’

  • 9

    6. Plasmid constructs and transfection

    To clone Pax6(+5a) and Pax6(–5a), these two isoforms were amplified using

    MLOY4 cDNA with the following primer sequences: forward sequence, 5’-

    GCGGTGAGCAGATGTGTG-3’, and reverse sequence, 5’-TCTCCTTCTC TCTTTACTGTAATC-3’.

    The PCR products were cloned into the T&A cloning vector (RBC Life Sciences,

    Real Biotech Corporation, Taiwan) and subcloned into a pcDNA3.1 mammalian

    expression vector (Invitrogen, Grand Island, NY, USA) after digestion with HindIII

    ((Promega, Madison, WI, USA) restriction enzyme to generate expression

    constructs (i.e., pcDNA3.1-Pax6(+5a) and pcDNA3.1-Pax6(-5a)). Cells were then

    seeded in 6-well plates and transfected with 0.8 µg of pax6 plasmid at 70-80%

    confluency using Lipofectamine PlusTM

    reagent (Invitrogen, Grand Island, NY,

    USA). At 48 h after transfection, the cells were processed to determine mRNA and

    protein expression levels of Pax6 and other osteocyte marker genes.

    7. Sost reporter plasmid construction

    The 1,811bp of the human Sost promoter (spanning nucleotides -1956 to -145bp

    of the Sost gene locus relative to the position (+1) of the initiation methionine for

    the Sost open reading frame) was amplified from liver cells with the following

    primer sequences forward sequence (5’-TCTCCCCCGGGTGTGGATCATTTAGAGTTCAAG-

    3’) and reverse sequence (5’-GCCCTAGATCTCCCAAAGACTTCTCCTCTAGCTC-3’). The

    promoter was cloned into a pGL3-basic luciferase reporter vector after digestion of

    the PCR product with SmaI and BglII (Promega, Madison, WI, USA) to construct

  • 10

    the pGL3-Sost promoter (wild type) plasmid. The primers used to generate

    mutations in the transcription factor binding site were as follows (mutated

    nucleotides in the primer sequence are indicated in bold letters): forward primer-5’-

    CTCTGGGTCACCTGGAAGTACAAACAGCAATTTGGAAGTTTGC-3’; and the reverse primer-5’-

    GCAAACTTCCAAATTGCTGTTTGTACTTCCAGGTGACCCAGAG-3’. A pGL3-Sost promoter

    (mut) plasmid was generated by site direct mutagenesis using expand long range

    dNTPack (Roche Diagnostics Corporation - Indianapolis, USA). PCR cycling

    conditions were as follows: initial denaturation at 950C for 4min followed by

    appropriate cycles (24cycles) at 950C for 45s (denaturation), 60

    0C for 45s

    (annealing) and 680C for 7min 15s (polymerization) and final extension at 68

    0 C for

    8 min. After PCR 5 l of the PCR reaction was digested with Dpn1 at 370 C for 1h

    and 5 l was transformed into DH5 alpha competent cells to prepare the mutant

    construct plasmids. All constructs sequence was confirmed by sequencing

    (Cosmogenetech, Seoul, South Korea).

    8. Transfection of cells with small interfering RNA

    Control siRNA (sc-37007; Santa Cruz Biotechnology Inc., CA, USA) and Pax6

    siRNA (sc-36196; Santa Cruz Biotechnology Inc., CA, USA) were used in this

    study. MLOY4 cells were seeded into 12-well plates and transfected with 50 pmoles

    of control siRNA or Pax6 siRNA using Lipofectamine RNAiMax reagent at 70%

    confluency (Invitrogen, Grand Island, NY, USA) according to the manufacturer’s

  • 11

    protocol. Cells were harvested 60 h after transfection. The siRNA activity was

    evaluated by measuring Pax6 mRNA suppression.

    9. Cell proliferation and cell number assay

    MLOY4 cells were cultured in 12-well plates and transfected with Pax6 siRNA.

    After 60 h, cell proliferation levels were determined using a cell counting kit-

    8(Dojindo Molecular Technologies Inc., Kumamoto, Japan) according to the

    manufacturer’s instructions. The cell number (viability) was determined by

    measuring lactate dehydrogenase (LDH) activity with a Cytotox 96 nonradioactive

    cytotoxicity assay (Promega, Madison, WI, USA). MLOY4 cells in 12-well plates

    were transfected with 0.6 µg of pcDNA3.1 empty vector, pcDNA3.1-Pax6(–5a) or

    pcDNA3.1-Pax6(+5a) plasmids using Lipofectamine PlusTM

    reagent. After 48 h, the

    effects of the Pax6 expression plasmids on cell proliferation and viability were

    studied.

    10. Western blot analysis

    Calvaria and longbone tissues of adult mouse were collected to check the

    expression of Pax6. To extract the proteins from tissues, tissue sections were

    washed with PBS and homogenized with lysis buffer (50mM Tris, pH 7.5;

    150mMNaCl; 1% Tween 20; 0.2%Nonidet P-40; 1mMphenylmethylsulfonyl

    fluoride; 500mM NaF; 1mM Na3VO4; 10 µg/ml aprotinin; 2 µg/ml pepstatin A; and

    10 µg/ml leupeptin). Proteins were extracted from the cells using the above lysis

  • 12

    buffer. The samples were centrifuged for 10 min at 12000 rpm at 40C and the

    supernatant was collected for western blot analysis. Protein concentration from each

    sample was determined by using Bradford reagent (Bio-Rad Laboratories, Hercules,

    CA, USA) according to manufacturer’s protocol. Equal amount of proteins, about

    25 µg were denatured at 1000C for 5 min. The extracted proteins were loaded and

    separated on 12% sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-

    PAGE) gels and transferred to polyvinylidene difluoride (PVDF) membranes

    (Millipore, Billerica, MA, USA). The membrane was blocked in TBST (TBS plus

    0.1% Tween-20), containing 5% skim milk for 1hr at room temperature and

    incubated overnight at 40C with an anti-Pax6 antibody (sc-53108; Santa Cruz

    Biotechnology Inc., CA, USA), anti-Sost antibody (R&D systems, Minneapolis,

    MN, USA), and anti-β-Actin antibody (sc-8432; Santa Cruz Biotechnology Inc., CA,

    USA). After washing, the membrane was incubated with HRP-conjugated anti-

    mouse, anti-goat immunoglobulin (Santa Cruz Biotechnology Inc., CA, USA) for 1

    h at room temperature. Immunoreactive proteins on the membrane were visualized

    with enhanced chemiluminescence (ECL) detection kits (Santa Cruz Biotechnology,

    CA, USA).

    11. Preparation of Wnt-3a conditioned medium

    Wnt-3a conditioned medium was obtained from established L-Wnt-3a stable cell

    lines (CRL-2647), which were purchased from ATCC (ATCC, Manassas, VA, USA).

    Wnt-3a conditioned medium was prepared according to manufacturer’s protocol

    (ATCC, Manassas, VA, USA).

  • 13

    12. Chromatin immunoprecipitation (Chip) assay

    Pax6 binding element on Sost promoter (~2kb) was analyzed by using r-vista

    software, which showed the putative Pax6 binding site (GGGAGTGCCAG) on the

    Sost promoter. This sequence was found to be a conserved region between mice and

    humans. For Chip assay, MLOY4 cells were cross-linked with 1% formaldehyde for

    10 min at room temperature. Cross linking was stopped by using 0.125M glycine,

    followed by washing with ice-cold PBS. Cells were resuspended in 1 ml Chip lysis

    buffer. After cell lysis, chromatin extracts were prepared and fragmented by

    sonication on ice. Sonication product (approximately 500 bp) was used as input

    after reverse cross-linking. Protein A/G beads (Santa Cruz Biotechnology Inc., CA,

    USA) were equilibrated with RIPA buffer and added to 50g of chromatin extract

    for preclearing. DNA-protein-antibody complexes were prepared using anti-Pax6

    antibody (sc-53108; Santa Cruz Biotechnology Inc., CA, USA) or normal mouse

    IgG antibody (sc-2026; Santa Cruz Biotechnology Inc., CA, USA) as a control

    following the Chip protocol (Abcam, Cambridge, MA, USA) with minor

    modifications. After precipitation of the chromatin, bound DNA was resuspended in

    water and used as a template for PCR using Sost Chip primers, forward 5’-

    TCTGAAAACCACAGCCTGAC-3’; and reverse 5’-GTTTCCTCACCCTCCTCCTC-3’ to amplify

    the conserved Pax6 binding site of mouse Sost promoter(-317 to -87 bp upstream to

    translation initiation site). For the control, Chip primers were designed in the other

    region of mouse Sost promoter (-1910 to -1676 bp), which do not contain binding

    element. The control Chip primers are as follows: forward primer 5’-

  • 14

    TCTCCCAAGTCTGGAGCAAT-3’, and reverse primer 5’-GCAGACAGAAGTCCCCTCAG-

    3’.

    13. Luciferase reporter assay for monitoring Sost promoter activity and Wnt

    signaling

    To study the role of Pax6 on Sost promoter activity MLOY4 cells were plated in

    24-well plate at a density of 55000cells/well. When cultures were 70-80% confluent

    cells were co-transfected using Lipofectamine PlusTM

    reagent with the indicated

    combinations of expression and Sost promoter plasmids. To study the role of Pax6

    in Wnt signaling, MLOY4 cells were plated in 12-well plate at a density of 1.1×105

    cells/well and subsequently co-transfected using Lipofectamine PlusTM

    reagent with

    the indicated combinations of expression and reporter plasmids. In the Wnt

    signaling study, the Super TOP Flash plasmid (S-Top) (Clontech Laboratories, Inc.,

    Palo Alto, CA, USA), which is driven by T-cell factor (TCF)/lymphoid enhancer

    factor (LEF)-binding elements and is responsive to Wnt signaling was co-

    transfected with Pax6 expression plasmids, and 24 h after transfection, the cells

    were treated with 100 µL of Wnt-3a conditioned medium. At 48 h after transfection,

    cell lysates were prepared, and luciferase assays were performed using the Dual-

    Luciferase Reporter Assay system (Promega, Madison, WI, USA) according to the

    manufacturer’s protocol. Assays were normalized for transfection efficiency by co-

    transfecting with 2.5 ng of pRL-tk renilla luciferase reporter plasmid.

  • 15

    14. Induction of Pax6 expression by Wnt-3a treatment

    MLOY4 cells were seeded in 12-well plates and treated with 100 µL of Wnt-3a

    conditioned media. After treatment Pax6 and Sost mRNA expressions were

    measured at different time intervals.

    15. Immunohistochemistry and HE staining

    For immunohistochemical staining, mandibles from mice at each stage were fixed

    with 4% paraformaldehyde in 0.01 M phosphate-buffered saline (PBS, pH 7.4)

    overnight at 4°C and demineralized with 10% ethylenediaminetetraacetic acid (pH

    7.4) for 2 weeks at 4°C. After being embedded in paraffin, these samples were

    sectioned to a thickness of 6 μm. Sections were then blocked in 3% hydrogen

    peroxide for 15 min. The tissue sections were boiled in 10 mM citrate buffer (pH

    6.0) for 10 min and cooled at room temperature for 20 min. The immunostained

    sections were then counterstained with hematoxylin. These sections were used for

    immunohistochemistry. Specimens were incubated with a primary mouse

    monoclonal antibody against Pax6 (Santa Cruz) at 4°C overnight. After washing

    with PBS, the specimens were incubated with a secondary antibody at room

    temperature. The specimens were visualized using a 3,3’-diaminobenzidine (DAB)

    reagent kit (Zymed laboratories, South San Fransisco, CA, USA).

  • 16

    16. Statistical analysis

    Statistical significance was evaluated using Sigma Plot10 software. The results

    were calculated and expressed as the mean ± standard deviation (SD). Statistical

    significance was determined using Student’s t-test. P-values ≤0.05 were considered

    to be statistically significant.

  • 17

    III. RESULTS

    1. Endogenous expression of Pax6 in various types of bone cells and tissues

    Endogenous expression of Pax6 in bone cells and tissues was studied. The

    expression of Pax6 was investigated in primary calvarial cells (early digestive

    fractions of calavarial tissue, which also called as primary-osteoblast cells

    (represented in materials and methods section)), MC3T3-E1 cells and osteocyte-like

    MLOY4 cells at the mRNA level. Pax6 mRNA was expressed at high levels in

    osteocyte-like MLOY4 cells compared to those in primary calvarial cells and

    MC3T3-E1 cells (Fig. 1A and 1B). Alternative splicing of Pax6 is known to result

    in two major Pax6 isoforms, both of which detected in MLOY4 cells (Fig. 1B). In

    addition, our western blot analysis demonstrated the in vivo expression pattern of

    Pax6 in calvaria and long bone tissues of adult mice (Fig. 1C). Pax6 expression at

    the protein level in MLOY4 cell lysates was confirmed by loading Pax6(–5a)- and

    Pax6(+5a)- overexpressing cell lysates. The Pax6(–5a) isoform was found in

    MLOY4 cells (Fig. 1D). To examine the expression of Pax6 during osteoblast

    differentiation, MC3T3-E1 cells were differentiated using osteoblast differentiation

    media. Differentiation of osteoblast cells was shown by ALP staining (Fig. 1E). The

    expression of Pax6 was gradually increased as the differentiation progressed (Fig.

    1F). Immunohistochemical studies showed the expression of Pax6 in osteoblasts

    and osteocytes in parietal bone region of calvaria sections at E16 and E18

    embryonic stages. Whereas the long bone sections of E16 and E18 showed

  • 18

    expression of Pax6 in both osteoblasts and osteocytes. Calvaria at E16 and E18

    stages showed comparatively higher expression than long bone (Fig. 2).

  • 19

    Figure 1. Endogenous expression of Pax6 in bone cells and tissues.

    A) RT-PCR analysis of Pax6 expression in mouse primary calvarial cells,

    MC3T3-E1 pre-osteoblast cells and osteocyte-like MLOY4 cells. (B)

    RT-PCR analysis of the endogenous expression of the two major

    i s o f o r ms o f P a x 6 [ P a x 6 ( + 5 a ) a n d P a x 6 ( – 5 a ) ] . T h e i r r e l a t i v e

    expressions are shown in the bar graph [Pax6–5a (black bar), Pax6+5a

    (gray bar)]. (C) Endogenous Pax6 expression in calvaria and long bone

    tissues was analyzed with western blotting and normalized to beta-actin

    expression. (D) Endogenous expression levels of Pax6 in MLOY4 lysates

    were compared with the overexpression of Pax6(–5a) and Pax6(+5a). (E)

    MC3T3-E1 cells differentiation was confirmed by ALP staining . (F)

    Pax6 expression was increased after induction of MC3T3-E1 cells with

    differentiation medium.

  • 20

    Figure 2. Immunohistochemical analysis of endogenous Pax6 expression.

    Immunohistochemical analysis of endogenous Pax6 expression in the calvaria

    and long bone tissues at embryonic stage 16 and 18. Brown color stained

    positive cells for Pax6 are indicated by arrows in each embryonic stages.

    Osteoblasts are indicated by red arrows, osteocytes are indicated by black

    arrows. Scale bar=50µm.

  • 21

    2. Effect of Pax6 on Wnt inhibitor Sost and other osteocyte markers

    The effect of Pax6 on osteocyte marker genes was studied via the knockdown or

    overexpression of Pax6 in MLOY4 cells. The gel images and semi-quantitative RT-

    PCR and real-time PCR data showed a significant knockdown of Pax6 (Fig. 3A and

    3B). Knockdown of Pax6 significantly decreased the expression of Sost (Fig. 3A

    and 3C). In contrast, the overexpression of Pax6(–5a) and Pax6(+5a) in MLOY4

    cells resulted in an significant increase in the expression of Sost, which is a mature

    osteocyte marker gene; meanwhile, there were no changes in the other marker genes,

    as observed with RT-PCR and real-time PCR (Fig. 3D-3G). Together, these results

    suggest that osteocyte-specific expression of Pax6 might be involved in the

    transition of pre-osteocytes to mature osteocytes and may also play an important

    role in inhibiting Wnt signaling through enhanced Sost expression. In addition to

    these results, studied the effect of Pax6 on the proliferation and cell number

    (viability) of MLOY4 cells. Present study showed that neither knockdown nor

    overexpression of Pax6 in MLOY4 cells had a significant effect on proliferation or

    cell number (Fig. 4A-4D).

  • 22

    Figure 3. The effects of Pax6 on osteocyte marker genes.

    The expression of osteocyte marker genes was analyzed with RT-PCR or real-time

    PCR. (A-C) Effect of siPax6 on osteocyte marker genes. RT-PCR and real-time

    PCR analysis showed decreased expression of Sost and increased expression of

    DMP-1 and E11 in siPax6 transfected cells compared to control siRNA transfected

    cells. (D-G) Effect of Pax6 overexpression on osteocyte marker genes. RT-PCR and

    real time PCR analysis showed significantly increased expression of Sost by Pax6

    overexpression compared to empty vector transfection.

  • 23

    Figure 4. Effect of Pax6 on proliferation and Cell viability.

    (A-B) Effect of siPax6 on cell proliferation and cell number. There is no effect on

    cell proliferation and cell number in cells treated with siPax6 compared to control

    siRNA. (C) Represents the effect of overexpression of Pax6(+5a) or Pax6(–5a)

    expression plasmids on cell proliferation compared to empty vector transfection.

    (D) Represents the effect of transient transfection of Pax6 expression plasmids on

    cell number compared to control vector. *Indicates a p-value ≤0.05 relative to the

    control.

  • 24

    3. Effect of Pax6 on Sost promoter activity in MLOY4 cells

    Pax6 binding site on the Sost promoter (~2kb) was found by insilico analysis (Fig.

    5A and 5B). This was further confirmed by Chip-PCR analysis. Pax6-bound

    chromatin was isolated from MLOY4 cells and immunoprecipitated with anti-Pax6

    antibody. The Pax6-bound DNA fragment shows amplification with specific primer

    set (Fig. 6A), whereas the control primers designed in the other region shows no

    amplification (Fig.6B). This result confirmed the specificity of Pax6 binding site on

    the Sost promoter. Since found that Pax6 regulates the expression of the Sost gene,

    further investigated the possible mechanism of Pax6 on Sost expression. To confirm

    the regulatory mechanism, 1,811bp of the human Sost promoter containing the

    Pax6-binding site (-1956 to -145 bp) was cloned into a pGL3-basic luciferase

    reporter vector (Fig. 7) by digestion with SmaI and BglII restriction enzymes, and

    evaluated the ability of this promoter fragment to drive luciferase expression in

    MLOY4 cells. Co-transfection of Pax6(+5a) or Pax6(–5a) plasmids with the wild

    type and mutant pGL3-Sost-promoter vectors led to a significant increase in

    promoter activity after overexpression of Pax6 in the wild type promoter and a

    decrease in activity in the mutant promoter in the presence of both isoforms of Pax6

    (Fig. 8). Since the Pax6 could not bind to the mutated region of the promoter, hence

    there was a significant decrease of reporter activity after overexpression of both

    forms of Pax6 compared to the wild type promoter (Fig. 8). This result suggested

    that Pax6 regulates Sost expression by binding to its promoter region specifically.

  • 25

    Figure 5. The Sost promoter sequence analysis (2kb region).

    (A) Sequence analysis of the 2 kb upstream regulatory region of human Sost

    promoter from transcription start site (2kb region shown here). Putative Pax6

    conserved binding site was represented in blue colour. The primers used for cloning

    of Sost promoter were represented in brown colour. (B) The conserved Pax6

    binding site.

  • 26

    Figure 6. Chip PCR analysis in MLOY4 cells.

    A) PCR amplification was performed by using Pax6 binding element region specific

    primers (primers indicated in blue colour) and B) negative control primers (primers

    indicated in violet colour) as described in the materials and methods section. This

    result confirms the specificity of Pax6 binding element on Sost promoter.

  • 27

    Figure 7. Schematic diagram of the Sost promoter-reporter construct.

    The Sost promoter region was cloned into SmaI and BglII sites in pGL3-basic

    vector (Promega) to make the Sost promoter-reporter construct.

  • 28

    Figure 8. The effect of Pax6 on Sost promoter activity.

    MLOY4 cells were transfected with the indicated combinations of the wild type and

    mutant Sost promoter and Pax6 expression plasmids. After 48 h, the relative

    luciferase activity was measured. *Indicates a p-value ≤0.05 relative to the control.

  • 29

    4. Effect of Wnt-3a on Pax6 mRNA expression in MLOY4 cells

    Sost is a well known negative regulator of Wnt signaling. Because Pax6

    regulates Sost expression, then investigated the effect of Wnt-3a on Pax6 mRNA

    expression in MLOY4 cells. Interestingly, found that Wnt-3a stimulated the

    expression of Pax6 mRNA at different time points, which further stimulated the

    expression of Sost mRNA (Fig. 9).

    Figure 9. Effect of Wnt3a on Pax6 mRNA expression.

    MLOY4 cells were treated with Wnt-3a conditioned media (100 µL/well), and the

    effect on Pax6 and Sost mRNA expression was measured at different time points

    with RT-PCR.

  • 30

    5. Effect of Pax6 on Wnt signaling in MLOY4 cells

    In addition, studied the effects of Pax6 on the S-Top reporter vector, which is

    driven by T-cell factor (TCF)/lymphoid enhancer factor (LEF)-binding elements

    and is responsive to Wnt signaling. However, the co-expression of the S-Top

    plasmid with Pax6 isoforms showed an inhibition of Wnt/β-catenin signaling in

    MLOY4 cells (Fig. 10A). Here also showed overexpression of pax6 and its effect

    on Sost expression in protein level through western blot analysis (Fig. 10B). These

    results suggested that Pax6 plays an important role in the inhibition of Wnt

    signaling mediated by the Sost gene in MLOY4 cells. Additionally, found that Wnt-

    3a enhanced Pax6 expression. In turn, overexpression of Pax6 inhibited Wnt

    signaling by increasing the expression of Sost, which is a potent inhibitor of Wnt

    signaling. Pax6 expression was increased according to further differentiation of

    osteoblasts. The role of Pax6 was represented schematically (Fig. 11).

  • 31

    Figure 10. Effect of Pax6 on Wnt signaling.

    A) MLOY4 cells were transfected with the indicated combinations of the Super

    TOPFlash reporter DNA and Pax6 expression plasmids. After 48 h, the relative

    luciferase activity was measured. *Indicates a p-value ≤0.05 relative to the control.

    B) Pax6 expression was confirmed by western blot analysis.

  • 32

    Figure 11. Schematic representation of the role of Pax6 in inhibiting Wnt

    signaling through Sost.

  • 33

    IV. DISCUSSION

    Canonical Wnt/β-catenin signaling plays important role in skeletal development,

    tissue regeneration and bone formation33

    . Postnatally, it also regulates bone

    homeostasis and bone mass34

    . Sost encodes sclerostin, which is specifically

    expressed by osteocytes and acts through a network of osteocyte canaliculi to reach

    the surface of the bone35,36

    . Sclerostin inhibits canonical Wnt signaling by binding

    to LRP5,637

    .

    In our GEO analysis, Pax6 was found to be a transcription factor that was

    upregulated with the Sost gene (data not shown). Pax6 acts as an important

    regulator in the development of the eye and central nervous tissues16,17

    . Pax6 is

    abundantly expressed in developing eyes, the forebrain, the ventral spinal card and

    the endocrine pancreas8. Homozygous mutant mice died at E6 to E8 before somite

    formation. Pax6 heterozygote mice have a small body size17

    . A Pax6-homozygous

    human fetus with aniridia showed an absence of nasal bones and defects in parietal

    bones at necropsy38

    . However, the role of Pax6 in bone has never been studied.

    Therefore, this study hypothesized that Pax6 might play an important role in bone

    development.

    In this paper, showed that compared with the expression of Pax6 in cultured

    primary calvarial cells or established MC3T3-E1 preosteoblast cells, Pax6 was

    exclusively expressed in MLOY4 osteocyte-like cells (Fig. 1A). Two major

    isoforms of endogenous Pax6 [Pax6(–5a) and Pax6(+5a)] were detected at the

    mRNA level in bone cells (Fig. 1B). However, this study showed that Pax6(–5a)

  • 34

    was the major Pax6 protein expressed in bone (Fig. 1D). Pax6 was expressed

    exclusively in the calvaria and long bone tissues at E16 and E18 (Fig. 2). Pax6 has

    been reported to be essential for the proliferation and the pluripotency of retinal

    progenitors39

    . The proliferation and apoptosis of human retinoblastoma cells are

    regulated by Pax640

    . To evaluate the role of Pax6 in bone, overexpression and

    knock down of Pax6 in MLOY4 cells was performed. In this study, neither

    knockdown nor overexpression of Pax6 exerted any effects on the proliferation or

    survival of MLOY4 cells (Fig. 4A-D). Additionally, observed that the knockdown

    of Pax6 decreased the expression of Sost mRNA, which is a late osteocyte marker

    gene, and increased the expression of E11 and DMP1 (Fig. 3A-3C). After transient

    transfection with Pax6(+5a) or Pax6(–5a) DNA, the expression of Sost was

    significantly increased (Fig. 3D and 3E). The induction of Sost expression by Pax6

    was further supported by the observation of enhanced luciferase reporter activity

    flanked by the 1811 bp promoter region of the Sost gene after overexpression of

    each isoform of Pax6 (Fig. 8). In other studies, Pax6 was found to play an important

    role in lens fiber cell and neuronal cell differentiation41,42

    . In this study, observed

    that the gradual increase of Pax6 mRNA during the differentiation of pre-osteoblast

    cells into mature osteoblasts (Fig. 1E and 1F). All these data suggest that Pax6

    might play an important role in the differentiation of osteoblasts and osteocytes.

    In chicken telencephalic development, Wnt-3a induced the expression of the

    dorsal markers Pax6 and Ngn243

    . Moreover, Pax6 was shown to play an important

    role in lens morphogenesis through the inhibition of canonical Wnt/ β-catenin

  • 35

    signaling in the lens surface ectoderm28

    . In this study, also observed the induction of

    Pax6 expression in MLOY4 osteocyte-like cells after treatment with Wnt-3a (Fig.

    9). In addition, Wnt-3a/β-catenin reporter activity was significantly reduced by the

    overexpression of both Pax6(+5a) and Pax6(–5a) along with the co-transfection of

    the S-Top reporter vector in osteocyte-like MLOY4 cells (Fig. 10A). After transient

    transfection with Pax6(–5a) or Pax6(+5a) DNA, the expression of Sost increased

    significantly (Fig. 3D and 3E). Additionally, our in silico analysis showed that

    mouse and human Sost promoters exhibited a conserved, putative Pax6-binding

    element (GGGAGTGCCAG) at the 2kb upstream region of the Sost promoter.

    Overexpression of Pax6(+5a) or Pax6(–5a) along with the pGL3-Sost promoter (wt)

    and the pGL3-Sost promoter (mut) DNA revealed that mutant promoter activity was

    significantly lower than wild type promoter activity in the presence of both isoforms

    of Pax6 (Fig. 8). All of these findings suggested that Pax6 inhibits canonical Wnt

    signaling through the stimulation of Sost by binding to its promoter. Canonical Wnt

    signaling has been shown to stimulate osteoblast differentiation and inhibit the

    terminal differentiation of mature osteoblasts and apoptosis44

    . In the present study,

    knockdown of Pax6 increased the expression of E11 and DMP1, and decreased the

    expression of Sost. In addition, Pax6 expression was increased according to further

    differentiation of osteoblasts. Therefore, speculated that Pax6 may be an important

    transcription factor in stimulating further differentiation of mature osteoblasts to

    osteocytes. The potential role of Pax6 is schematically represented in Fig.10.

  • 36

    V. CONCLUSION

    The present study showed that the transcription factor Pax6 is expressed in the

    osteocyte-like MLOY4 cells, calvaria and long bone tissues. Pax6 expression was

    observed during the terminal differentiation of MC3T3E1- cells. Overexpression of

    Pax6 increased the expression of Sost, a late osteocyte marker gene. By direct

    binding to the putative Pax6 binding sequence of the Sost promoter region, Pax6

    suppressed the canonical Wnt signaling pathway. Further exploration of phenotype

    in osteocyte-specific Pax6 knockout mice could provide more definitive evidence of

    the role of Pax6 in differentiation of mature osteoblasts to osteocytes.

  • 37

    REFERENCES

    1. Yang Xi, karsenty G. Transcription factors in bone: developmental and

    pathological aspects. Trends in Mol med 2002;8:340-5.

    2. Dallas SL, Bonewald LF. Dynamics of the transition from Osteoblast to

    Osteocyte. Ann N Y Acad Sci 2010;1192:437-43.

    3. Lanyon LE. Osteocytes, strain detection, bone modeling and remodeling.

    Calcif Tissue Int 1993;53:102-7.

    4. Franz-Odendaal TA, Hall BK, Witten PE. Buried alive: How osteoblasts

    become osteocytes. Dev Dyn 2006;235:176-90.

    5. van Bezooijen RL, Roelen BA, Visser A, van der Wee-Pals L, de Wilt E,

    Karperien M, et al. Sclerostin is an osteocyte expressed negative regulator

    of bone formation, but not a classical BMP antagonist. J Exp Med

    2004;199:805-14.

    6. Semenov M, Tamai K, He X. SOST is a ligand for LRP5/LRP6 and a WNT

    signaling inhibitor. J Biol Chem 2005;280:26770-75.

    7. van Bezooijen RL, Bronckers AL, Gortzak RA, Hogendoorn PC, van der

    Wee-Pals L, Balemans W, et al. Sclerostin in mineralized matrices and van

    buchem disease. J Dent Res 2009;88:569-74.

  • 38

    8. De Crombrugghe B, Lefebvre V, Nakashima K. Regulatory mechanisms in

    the pathways of cartilage and bone formation. Curr Opin Cell Biol

    2001;13:721-7.

    9. Karsenty G, Wagner EF. Reaching a genetic and molecular understanding

    of skeletal development. Dev cell 2002;2:389-406.

    10. Komori T. Regulation of osteoblast differentiation by transcription factors.

    J Cell Biochem 2006;99:1233-9.

    11. Marie PJ. The molecular genetics of bone formation:implications for

    therapeutic interventions in bone disorders. Am J Pharmacogenomics 2001;

    1:175–87.

    12. Barrett T, Troup DB, Wilhite SE, Ledoux P, Rudnev D, Evangelista C, et al.

    NCBI GEO: mining tens of millions of expression profiles database and

    tools update. Nucleic Acids Res 2007;35:D760-5.

    13. Walther C, Gruss P. Pax6, a murine paired box gene, is expressed in the

    developing CNS. Development 1991;113:1435-49.

  • 39

    14. Hogan BL, Hirst EM, Horsburgh G, Hetherington CM. small eye (Sey): a

    mouse model for the genetic analysis of craniofacial abnormalities.

    Development 1988;103: 115-9.

    15. Kaufman MH, Chang HH, Shaw JP. Craniofacial abnormalities in

    homozygous Small eye (Sey/Sey) embryos and newborn mice. J Anat

    1995;186:607-17.

    16. Stoykova A, Fritsch R, Walther C, Gruss P. Forebrain patterning defects in

    small eye mutant mice. Development 1996;122:3453-65.

    17. Theiler K, Varnum DS, Stevens LC. Development of Dickie’s small eye, a

    mutation in the house mouse. Anat Embryol (Berl) 1978;155:81-6.

    18. Carriere C, Plaza S, Caboche J, Dozier C, Bailly M, Martin P, et al. Nuclear

    localization signals, DNA binding, and transactivation properties of quail

    Pax6 (Pax-QNR) isoforms. Cell Growth Differ 1995;6:1531-40.

    19. Jaworski C, Sperbeck S, Graham C, Wistow G. Alternative splicing of Pax6

    in bovine eye and evolutionary conservation of intron sequences. Biochem

    Biophys Res Commun 1997;240:196-202.

  • 40

    20. Kim J, Lauderdale JD. Analysis of Pax6 expression using a BAC transgene

    reveals the presence of a paired-less isoform of Pax6 in the eye and

    olfactory bulb. Dev Biol 2006;292:486-505.

    21. Tang HK, Singh S, Saunders GF. Dissection of the transactivation function

    of the transcription factor encoded by the eye developmental gene Pax6. J

    Biol Chem 1998;273:7210-21.

    22. Zhang W, Cveklova K, Oppermann B, Kantorow M, Cvekl A. Quantitation

    of Pax6 and Pax6(5a) transcript levels in adult human lens, cornea, and

    monkey retina. Mol Vis 2001;7:1-5.

    23. Berger J, Berger S, Tuoc TC, D'Amelio M, Cecconi F, Gorski JA, et al.

    Conditional activation of Pax6 in the developing cortex of transgenic mice

    causes progenitor apoptosis. Development 2007;134:1311-22.

    24. Cillo C, Faiella A, Cantile M, Boncinelli E. Homeobox genes and cancer.

    Exp Cell Res 1999;248:1-9.

    25. Haubst N, Berger J, Radjendirane V, Graw J, Favor J, Saunders GF, et al.

    Molecular dissection of Pax6 function: the specific roles of the paired

    domain and homeodomain in brain development. Development

    2004;131:6131-40.

  • 41

    26. Lauderdale JD, Wilensky JS, Oliver ER, Walton DS, Glaser T. 3’ deletions

    cause aniridia by preventing Pax6 gene expression. Proc Natl Acad Sci

    USA 2000;97:13755-9.

    27. Wodarz A, Nusse R. Mechanisms of Wnt signaling in development. Annu

    Rev Cell Dev Biol 1998;14:59-88.

    28. Machon O, Kreslova J, Ruzickova J, Vacik T, Klimova L, Fujimura N, et al.

    Lens morphogenesis is dependent on Pax6 mediated inhibition of the

    Canonical Wnt/beta-Catenin signaling in the lens surface ectoderm. Genesis

    2010;4:86-95.

    29. Johnson ML, Harnish K, Nusse R, Van Hul W. LRP5 and Wnt signaling: a

    union made for bone. J Bone Miner Res 2004;19:1749-57.

    30. Kato Y, Windle JJ, Koop BA, Mundy GR, Bonewald LF. Establishment of

    an osteocyte –like cell line, MLOY4. J Bone Miner Res 1997;12:2014-23.

    31. Ma Y, Wu X, Li X, Fu J, Shen J, Li X, Wang H. Corticosterone regulates

    the expression of neuropeptide Y and reelin in MLOY4 cells. Mol Cells

    2012;33:611-6.

  • 42

    32. Park SJ, Kim SJ, Rhee Y, Byun JH, Kim SH, Kim MH, et al. Fidgetin-like1

    gene inhibited by basic fibroblast growth factor regulates the proliferation

    and differentiation of osteoblasts. J Bone Miner Res 2007;22:889-96.

    33. Krishnan V, Bryant HU, Macdougald OA. Regulation of bone mass by Wnt

    signaling. J Clin Invest 2006;116:1202-9.

    34. Knothe Tate ML, Adamson JR, Tami AE, Bauer TW. The osteocyte. Int J

    Biochem Cell Biol 2004;36:1-8.

    35. Poole KE, van Bezooijen RL, Loveridge N, Hamersma H, Papapoulos SE,

    Lowik CW, et al. Sclerostin is a delayed secreted product of osteocytes that

    inhibits bone formation. FASEB J 2005;19:1842-4.

    36. Weidauer SE, Schmieder P, Beerbaum M, Schmitz W, Oschkinat H,

    Mueller TD. NMR structure of the Wnt modulator protein sclerostin.

    Biochem Biophys Res Commun 2009;380:160-5.

    37. Li X, Zhang Y, Kang H, Liu W, Liu P, Zhang J, et al. Sclerostin binds to

    LRP5/6 and antagonizes canonical Wnt signaling. J Biol Chem

    2005;280:19883-7.

  • 43

    38. Hodgson SV, Saunders KE. A probable case of homozygous condition of

    the aniridia gene. J Med Genet 1980;17:478-80.

    39. Hsieh YW, Yang XJ. Dynamic Pax6 expression during the neurogenic cell

    cycle influences proliferation and cell fate choices of retinal progenitors.

    Neural Dev 2009;4: 32.

    40. Bai SW, Li B, Zhang H, Jonas JB, Zhao BW, Shen L, et al. Pax6 regulates

    proliferation and apoptosis of human retinoblastoma cells. Invest

    Ophthalmol Vis Sci 2011;52:4560-70.

    41. Gotz M, Stoykova A, Gruss P. Pax6 controls radial glia differentiation in

    the cerebral cortex. Neuron 1998;21:1031-44.

    42. Shaham O, Smith AN, Robinson ML, Taketo MM, Lang RA, Ashery-Padan

    R. Pax6 is essential for lens fiber cell differentiation. Development

    2009;136:2567-78.

    43. Gunhaga L, Marklund M, Sjodal M, Hsieh JC, Jessell TM, Edlund T.

    Specification of dorsal telencephalic character by sequential Wnt and FGF

    signaling. Nat Neurosci 2003;6:701-7.

  • 44

    44. Manolagas SC. Birth and death of bone cells: basic regulatory mechanisms

    for the pathogenesis and treatment of osteoporosis. Endocr Rev

    2000;21:115-37.

  • 45

    ABSTRACT (IN KOREAN)

    Osteocyte에서 발현하는 Pax6에 의한 canonical wnt signaling 억제

    연세대학교 대학원 의과학과

    아지타 자미

    Pax6는 paired box-containing domain을 갖는 전사인자로서, 눈, 중추신경조

    직, 두개구조의 발달과정을 조절한다. 그러나 뼈에서는 아직까지 Pax6의 역할

    이 정확하게 알려지지 않았다. 본 연구에서는 성체 마우스의 두개관(calvaria)

    과 대퇴골, MLOY4 세포주에서 Pax6가 발현되며 canonical Wnt 신호전달체계

    가 억제됨을 확인하였다. 또한 Pax6는 두개관과 대퇴골의 E16, E18 단계에서

    모두 표현되었고, 대퇴골에서보다 두개관에서 Pax6의 발현이 더 높았다.

  • 46

    MLOY4 세포주에서 Pax6를 knockdown 하였을 때 세포증식에는 영향을 미치

    지 않았으나, late osteocyte marker인 Sost가 현저히 감소함을 확인하였다. 또한,

    Pax6를 과발현하였을 때에는 E11및 DMP1표현의 변화는 관찰되지 않았지만,

    Sost의 표현이 증가되었고, Sost에 의한 canonical Wnt 신호전달체계가 억제됨

    을 관찰할 수 있었다. 본 연구에서는 이것을 증명하기 위해 Pax6가 Sost의 특

    정 부위 (-GGGAGTGCCAG-)에 결합함으로써 Sost의 전사과정을 조절함을 보

    였다. 이로써 Pax6가 뼈에서도 canonical Wnt신호전달체계를 조절하고 골세포

    (osteocyte)의 분화에 중요한 역할을 함을 알 수 있었다.

    핵심되는 말 : Pax6, Osteocyte-like MLOY4 cells, MC3T3E1 cells, Sost, Wnt signaling

  • 47

    PUBLICATION LIST

    1. Narinder P singh, Gopal K Ingle, Vinay K Saini, Ajita Jami, Pankaj

    Beniwal, Madan Lal , Gajender S Meena. Prevalence of low glomerular

    filtration rate, proteinuria and associated risk factors in North India using

    Cockcroft-Gault and Modification of Diet in Renal Disease equation: an

    observational, cross-sectional study. BMC Nephrology 2009, 10:4.

    2. Kyoung Min Kim, Su Jin Park, Seung-Hyun Jung, Eun Jin Kim,

    Jogeswar Gadi, Ajita Jami, Yumie Rhee, Cheol-Hee Kim, Sung-Kil Lim.

    miR-182 is a Negative regulator of osteoblast proliferation,

    differentiation, and skeletogenesis through targeting FoxO1. JBMR 2012;

    27(8):1669-1679.

    3. Ajita Jami, Jogeswar Gadi, Min Jung Lee, Eun Jin Kim, Mi Jeong Lee,

    Han-Sung Jung, Hong-Hee Kim, and Sung-Kil Lim. Pax6 expressed in

    osteocytes inhibits canonical wnt signaling. Mol Cells 2013 April; 35(4):

    305-12.