ROLES OF MYOCARDIN RELATED TRANSCRIPTION FACTORS IN

MUSCLE AND BRAIN

APPROVED BY SUPERVISORY COMMITTEE

Eric N. Olson, Ph.D.

Melanie Cobb, Ph.D.

James Stull, Ph.D.

Jane Johnson, Ph.D.

To my beloved family…

Acknowledgements

I have had the opportunity to work with and discuss science with many great people in the Olson lab.

First, I would like to thank my mentor Dr. Eric Olson for giving me the opportunity to be part of his research team. I will always be grateful for the training I received in his lab and his support throughout my graduate career.

I would like to thank Dr. Rhonda Bassel-Duby for her everyday support.

I would like to thank my thesis committee members, Drs. Melanie Cobb, James Stull, and

Jane Johnson for their advice during my training.

I would like to thank to Dr. James Richardson, John Shelton, and the histology core members for their help on histological sections and discussion. I would also like to thank members of the Department of Molecular Biology, especially Jose Cabrera for graphics, Jennifer

Brown for help with manuscripts and travel, and Wanda Simpson for help with scheduling meetings. Most of this work would not be possible without the help of Xiaoxia Qi in performing targeting in ES cells, John McAnally for generating transgenic mice, and Cheryl Nolen for technical support.

In addition, I would like to thank all past and present members of the Olson lab who have facilitated my scientific and nonscientific experiences in Dallas.

I am particularly grateful to Aaron Johnson, Michael Haberland, and Rusty Montgomery for always being there for me. I have been very fortunate to have them guide my research experience, support me through everyday life, and influence the way I approach science and life.

I also would like to thank Andrew Williams, Gaile Vitug, Michele Carrer, Yuri Kim, Chris Davis,

Nik Munshi, Ana Barbosa, Eva van Rooij, Jiyeon Oh, Peng Yi, Esther Creemers, Shusheng

Wang, Ning Liu, Viviana Moersi, Lillian Sutherland, Svetlana Bezprozvannanya, Eric Small,

Mi-Sung Kim, Chad Grueter, and Kunhua Song for being awesome lab mates.

I am also greatly indebted to numerous people back in Lebanon for all their efforts to support me throughout my years in Dallas.

I am grateful for my two mentors at the American University of Beirut, Drs. Marwan El-Sabban and Rabih Talhouk, who guided my first training in science, encouraged me to pursue my career, and have been extremely supportive ever since.

Finally, I would like to express my gratitude and love to my family and my wonderful parents. I am extremely fortunate to have them and indebted for their never ending love, support, and guidance. I would like to thank my mom and dad for the huge ambition they implanted in me and for working very hard to provide me with the best opportunities to follow it.

ROLES OF MYOCARDIN RELATED TRANSCRIPTION FACTORS IN

MUSCLE AND BRAIN

by

Mayssa H. Mokalled

DISSERTATION

Presented to the Faculty of the Graduate School of Biomedical Sciences

The University of Texas Southwestern Medical Center at Dallas

In Partial Fulfillment of the Requirements

For the Degree of

DOCTOR OF PHILOSOPHY

The University of Texas Southwestern Medical Center at Dallas

Dallas, Texas

September, 2009

Copyright

by

Mayssa H. Mokalled, September 2009

All Rights Reserved

ROLES OF MYOCARDIN RELATED TRANSCRIPTION FACTORS

IN MUSCLE AND BRAIN

Mayssa H. Mokalled

The University of Texas Southwestern Medical Center at Dallas, 2009

Mentor: Eric N. Olson, Ph.D.

Partnerships between DNA binding transcription factors and transcriptional cofactors govern gene transcription in various developmental and tissue contexts, particularly during cardiovascular and neuronal development. This dissertation aims at studying the in vivo relevance of the partnership between Serum response Factor (SRF) and its Myocardin Related Transcription Factor (MRTF) coactivators during development. I present here my studies on the functions of MRTFs during brain and muscle development.

First, I show that MRTF-A and -B redundantly control neuronal migration and neurite outgrowth during brain development. Conditional deletion of these in the mouse brain disrupts the formation of multiple brain structures, reflecting a failure in neuronal actin polymerization and cytoskeletal assembly. I also describe a previously unrecognized role for the MRTF/SRF pathway in the regulation of the Pctaire-1/Cdk5 cascade to govern actin dynamics. I conclude that MRTFs function as essential

vii

coregulators of SRF to control actin dynamics during neuronal development via the

Cdk5/Pctaire-1 kinase cascade.

I also explore the role of MRTF-A and -B in cardiac development and function.

Ablation of these MRTF genes in the embryonic heart causes` a range of cardiac defects, reflecting the sensitivity of cardiac function to MRTF gene dosage. Moreover, I show that the gene encoding Pctaire-1 kinase, whose functions in the heart are unknown, is also a target of MRTF/SRF signaling and a regulator of sarcomere assembly in the heart.

Furthermore, by creating mice lacking the Myocardin related factor MASTR, I explore the in vivo developmental functions of MASTR. Germline deletion of MASTR alone does not cause any obvious defects in mice. However, deletion of MASTR in an

MRTF-A null background causes perinatal lethality, which appears to be due to defective skeletal muscle growth and development.

Thus, the results of my thesis research demonstrate that MRTFs are essential regulators of multiple developmental processes in brain, heart, and skeletal muscle. At the cellular level, MRTFs are essential regulators of the actin cytoskeleton. Disruption of

MRTF functions, whether in neurons or in muscle cells, causes major cytoskeletal defects that impair brain and muscle development and function.

viii

Table of Contents

Title………………………………………………………………………………………... i Dedication………………………………………………………………………………… ii Acknowledgements……………………………………………………………………….iii Abstract………………………………………………………………………………….. vii Table of Contents………………………………………………………………………… ix List of Publications………………………………………………………………………. xi List of Figures………………………………………………………………………….... xii List of Abbreviations…………………………………………………………………….xiv Chapter I. Introduction……………………………………………………………………1 Transcriptional cofactors in development …………………………..……………2 Regulation of SRF activity in various cell types..………………………………...3 Multifunctionality of SRF……………...……………………...………………….5 Domain structure of MRTFs……...………...…………………………………….7 Implication of MRTFs in multiple disease processes.…………………………...11 Myocardin and MASTR, coactivators of SRF and MEF2 in muscle cells...... 12 Contribution of MRTF-A and -B to SRF signaling……………………………...14 Putative MRTF functions in brain and muscle..………….…..………………….15 Chapter II. MRTFS regulate the Cdk5/Pctaire-1 kinase cascade to control neurite outgrowth and neuronal migration during brain development…………………..18 Abstract…...……………………………………………………………………..19 Introduction……………………………………………………………….……..20 Results…………………………………………………………………...……….23 Generation of mice lacking MRTF-A and -B in the brain………………...…….. 23 MRTF bdKO mice have severe brain abnormalities …………………………... 26 Defective neuronal migration along the rostral migratory stream……………….26 Defective neurite outgrowth in MRTF bdKO neurons..…………………………31 Pctaire-1 is a novel MRTF/SRF target gene………………………………..…… 33 Regulation of actin dynamics by MRTFs……………………………………….. 36 Discussion……………………………………………………...………………...39

ix

Functions of MRTFs in vivo………………………………………………………... 41 MRTFs as regulators of the actin cytoskeleton dynamics……………………….41 Identification of Pctaire-1as a novel MRTF/SRF target gene in the brain……... 43 Methods…………………………………………………………………………. 44 Chapter III. A threshold of MRTFs is required for cardiac development and function….50 Abstract……………………………………………………………………...…... 51 Introduction…………………………..…………………………………………. 52 Results……………………………………...……………………………………. 53 Generation of mice lacking MRTF-A and -B in the heart….…………………… 53 Phenotype of mice lacking MRTF-B in MRTF-A null background……………..54 Defective sarcomere arrangement in MRTF cdKO hearts……………………… 56 Molecular dissection of the MRTF cdKO phenotype………………………….. 56 Pctaire-1 in cardiac development …………………..…………………………… 59 Discussion……………………………………………………………………….. 64 Redundant functions of MRTFs in the heart……….…………………………… 64 MRTFs in SRF signaling……….……………………………………………….. 66 Functions of Pctaire-1 in cardiac development……..…………………………… 66 Methods…………………………………………………………………………..68 Chapter IV. MASTR cooperates with MRTF-A to control skeletal muscle development…………………………………………………………………….. 71 Abstract……………………………………………………………………...…... 72 Introduction…………………………..…………………………………………. 73 Results……………………………………...……………………………………. 75 Expression analysis of MASTR…………...……………………………………. 75 Generation of MASTR knockout mice..…...……………………………………. 75 Generation of MASTR/MRTF-A double knockout mice.………………………. 76 Discussion……………………………………………………………………….. 79 Methods…………………………………………………………………………..80 Chapter V. Summary and Future Directions…………………………………………… 82 Summary………………………………………………………………………… 83 Future directions………………………………………………………………… 71

x

Bibliography……………………………………………………………………………...86

xi

List of Publications

Mokalled, M.H., Richardson, J.A., and Olson E.N. A Threshold of Myocardin Related Transcription Factors in Cardiac development and Function. Manuscript in preparation.

Mokalled, M.H., Johnson, A., Kim, Y., Oh, J., and Olson, E.N. Myocardin Related Transcription Factors Regulate the Cdk5/Pctaire-1 Kinase Cascade to Control Neurite Outgrowth, Neuronal Migration and Brain Development. Submitted.

Haberland, M., Mokalled, M.H., Montgomery, R.L., and Olson, E.N. (2009). Epigenetic Control of Skull Morphogenesis by Histone Deactelylase 8. Genes Dev. 23(14),1625- 1630.

Haberland, M., Johnson, A., Mokalled, M.H., Montgomery R.L., and Olson, E.N. (2009). Genetic Dissection of Histone Deacetylase Requirement in Tumor Cells. Proc Natl Acad Sci USA. 106(19), 7751-7755.

xii

List of Figures

Figure 1.1 Domain structure of the Myocardin Related Transcripotion Factors………… 8 Figure 1.2 Activation of MRTF in response to extracellular signaling…………………..10 Figure 2.1 Generation of MRTF brain double knockout (bdKO) mice...………………...24 Figure 2.2 Neuroanatomical defects in MRTF bdKO mice………………………………27 Figure 2.3 Deletion of MRTF-A and -B in mouse embryonic fibroblasts (MEFs).……...29 Figure 2.4 Defective neuronal cell migration in MRTF bdKO mice...…………………...30 Figure 2.5 Decreased neuronal projections in MRTF bdKO brains ..…………………….32 Figure 2.6 Aberrant expression of Actin and Gelsolin in MRTF bdKO mice.……………34 Figure 2.7 Identification of Pctaire-1 as a novel MRTF/SRf target gene………………....35 Figure 2.8 Regulation of actin dynamics by the MRTF/SRF pathway…………………....37 Figure 2.9 Proposed model for the regulation neuronal migration and neurite outgrowth by the MRTF/SRF pathway..……….………………..……………………….40 Figure 3.1 Genration of MRTF cdKO mice…………………..…………………………...55 Figure 3.2 A range of cardiac defects upon MRTF deletion in the heart…….……………57 Figure 3.3 Defects in sarcomere arrangement in MRTF bdKO hearts ……………………58 Figure 3.4 Dysregulation of sarcomere components in MRTF cdKO hearts.……………..60 Figure 3.5 Pctaire-1 is a mediator of MRTF/SRF signaling in the heart.....……………….61 Figure 3.6 Pctaire-1 regulates cardiac development in vivo…..……. …………….………..63 Figure 3.7 Model of MRTF function in the heart …………………………………………65 Figure 4.1 Expression analysis of MASTR…...…………………………………………...74 Figure 4.2 Generation of MASTR knockout mice...…………………………………..…..77 Figure 4.3 Defective skeletal muscle development in MRTF-A/MASTR dKO mice..…...78

xiii

List of Abbreviations bdKO brain double knockout BrdU bromodeoxyuridine cdKO cardiac double knockout Cdk5 cyclin dependent kinase 5 cDNA complementary DNA dKO double knockout DNA deoxyribonucleic acid ES embryonic stem F floxed GAPDH glyceraldehyde-3-phosphate dehydrogenase H&E hematoxylin and eosin LimK Lim Kinase Luc luciferase MADS MCM1, agamous, deficiens, SRF MASTR MEF2 activating SAP transcriptional regulator MEF2 myocyte enhancer factor 2 MRTF myocardin related transcription factor P postnatal day PBS phosphate buffered saline PCR polymerase chain reaction Q-PCR quantitative PCR RNA ribonucleic acid RT-PCR reverse transcriptase-polymerase chain reaction SRF serum response factor Tuj1 tubulin J1 UTR untranslated region

xiv

Chapter I

Introduction

Transcriptional cofactors in development

Gene transcription is a multistep process that requires concerted activation of a network of transcription factors and cofactors. The expression and activity of DNA binding transcription factors is tightly regulated to control the onset, localization, and level of expression of their target genes. The activity of transcription factors depends largely on the expression and activity of transcriptional coregulators. Transcriptional cofactors assemble with their transcription factor partners into activator or repressor complexes that alter the chromatin structure of a target gene to enhance or repress its transcription.

Cofactor mediated modulation of gene expression thus allows dynamic responsiveness of a transcription factor to multiple signaling events. It also provides tissue and developmental specificity to widely expressed transcription factors. Therefore, it is not surprising that transcriptional cofactors evolved as primary targets of developmental or physiological signals (Liu and Olson, 2006; Spiegelman and Heinrich, 2004).

Several transcription factor and partnerships have been described in various developmental and tissue contexts, particularly during cardiovascular and neuronal development. In the brain, the steroid receptor coactivators (SRC-1, -2, -3), which activate estrogen, glucocorticoid, and thyroid hormone signaling, are differentially expressed in various brain structures and required for normal neuronal development and functions (Alonso et al., 2009; Apostolakis et al., 2002; Nishihara et al., 2003). Similarly, association between the GATA transcription factors and their Friend of GATA (FOG) coregulators in the heart illustrates the biological relevance of transcriptional coactivation during cardiogenesis. Germline deletion of FOG-2 for instance causes cardiac malformations and embryonic lethality by midgestation, demonstrating that GATA

2

functions rely on its coactivator activity (Spiegelman and Heinrich, 2004; Tevosian et al.,

2000). Similarly, other cofactors such as CAMTA and TAZ partner with the cardiac transcription factors Nkx2.5 and Tbx5, respectively, to regulate gene expression during cardiac development and remodeling (Liu and Olson, 2006).

This dissertation aims at studying the in vivo relevance of yet another partnership between the DNA binding Serum Response Factor (SRF) and its Myocardin Related

Transcription Factor (MRTF) coactivators, particularly during neuronal and muscle development.

Regulation of SRF activity in various cell types

SRF, a member of the MADS (MCM1, Agamous, Deficiens, SRF) box family of transcription factors, binds to a consensus DNA sequence (CC(A/T)6GG) called a CArG box. The widely expressed SRF transcription factor orchestrates multiple programs of gene expression, ranging from cell growth to muscle differentiation. Several regulatory mechanisms exert tight control on SRF activity to ensure context and cell specific expression of SRF target genes. These include regulation of the expression, alternative splicing, and DNA binding of SRF. The activity of SRF is also modulated by remodeling the chromatin structure of CArG boxes and adjacent sequences and by association of SRF with cell restricted cofactors (Miano, 2003; Parmacek, 2007; Pipes et al., 2006).

Two families of coactivators, Ternary Complex factors (TCFs) and Myocardin

Related Transcription Factors (MRTFs), share structurally related SRF binding domains that bind the same region of SRF. MRTFs and TCFs modulate the transcriptional activity of SRF in response to signaling from the Rho GTPase and Mitogen Activated Protein

3

Kinase (MAPK) pathways, respectively. Upon activation of MAPK signaling, members of the TCF family of Ets domain proteins (Elk-1, SAP-1 and SAP-2) bind to Ets motifs adjoining the CArG box of some immediate-early genes, such as c-fos, to drive their transcription (Posern and Treisman, 2006). MRTFs, on the other hand, are either constitutively active, in the case of Myocardin, or activated by Rho signaling, in the case of MRTF-A and -B. MRTFs and TCFs are recruited to the SRF/DNA complex in a mutually exclusive manner. Hence, it is not surprising that, when assessed by chromatin immunoprecipitation assays, SRF target genes were grouped into either MRTF or TCF dependent genes (Miralles et al., 2003).

The mechanisms that dictate differential activation of different SRF target genes and the in vivo relevance of MRTF versus TCF dependent coactivation of SRF are still to be investigated. These mechanisms have been studied in smooth muscle cells, which are capable of switching between a proliferative synthetic state and a differentiated contractile state in response to various signaling pathways. In response to growth signals,

Elk-1, a member of the TCF family of cofactors, promotes cell proliferation and inhibits myogenesis by displacing the muscle restricted MRTF member, myocardin, from SRF

(Wang et al., 2004). However, this mechanism was not extended to other cell types that do not exhibit the dynamic switching between proliferation and differentiation gene programs.

In addition to the MRTF and TCF cofactors, SRF interacts with multiple other transcription factors. GATA and Nkx2.5 transcription factors bind SRF and form complexes with their own adjacent DNA binding sites to enhance SRF mediated gene transcription. On the other hand, the LIM-only protein FHL2 and the Heart-enriched

4

homeodomain only cofactor HOP negatively regulate SRF activity (Posern and Treisman,

2006).

The activity of SRF is also regulated by phosphorylation. The Myotonic

Dystrophy Protein Kinase (DMPK) and Protein Kinase Cα (PKCα) phosphorylate SRF within the MADS box at Thr159 and Thr162, respectively, mediating switching between proliferation and differentiation gene programs in smooth muscle cells (Iyer et al., 2003;

Iyer et al., 2006). Phosphorylation of SRF at Thr162 alters the conformation of the

SRF/DNA complex, destabilizing its binding to MRTFs, without affecting TCF binding

(Iyer et al., 2006).

Multifunctionality of SRF

Mouse embryos homozygous for an SRF null mutation fail to generate mesoderm and die at gastrulation (Arsenian et al., 1998). However, tissue specific deletion of SRF has revealed multiple roles for this transcription factor in brain, muscle, heart, and liver development (Charvet et al., 2006; Latasa et al., 2007; Miano et al., 2004; Niu et al.,

2005; Parlakian et al., 2005).

Conditional deletion of SRF in the brain disrupts the actin cytoskeleton impairing neurite outgrowth, axon guidance in response to ephrin and semaphorin signaling, and directional migration of neurons from the subventricular zone to the olfactory bulb

(Alberti et al., 2005; Knoll et al., 2006). Deletion of SRF in the brain also disrupts the formation of hippocampal neuronal circuitry resulting in defective memory formation.

SRF null CA1 pyramidal neurons, which exhibit normal cellular morphology and basal excitatory transmission, display a diminished long-term synaptic potentiation (LTP) and

5

impaired induction of long-term synaptic depression (LTD), indicating that SRF is required for plastic modification of synaptic strength in response to activity in the adult brain (Etkin et al., 2006; Ramanan et al., 2005).

Several cardiac specific SRF knockouts have been characterized to date. Deletion of SRF in the embryonic heart using beta-MHC-Cre transgenic mice impairs cardiac differentiation and maturation and disrupts the expression of multiple regulators of cardiac development, such as Nkx2.5 and GATA4, resulting in embryonic lethality between E10.5 and E13.5 (Parlakian et al., 2004). Ablation of SRF using alpha-MHC-

Cre mice results in cardiac insufficiency and reduced cellularity during cardiogenesis accompanied by reductions in SRF, ANF, and cardiac, skeletal, and smooth muscle alpha-actin transcripts (Niu et al., 2005). Deletion of SRF in the adult heart using a heart- specific tamoxifen-inducible Cre recombinase demonstrates its requirement for adult cardiac function and integrity. Following tamoxifen treatment, cardiac defects developed gradually from impairment of left ventricular function to dilated cardiomyopathy and eventually heart failure, with disruption of cardiomyocyte cytoarchitecture and disorganization of intercalated discs (Parlakian et al., 2005).

SRF has also been shown to be required for smooth and skeletal muscle development. Deletion of SRF in cardiomyocytes and smooth muscle (SM) cells using

SM22-Cre transgenic mice causes multiple defects in the heart and vasculature accompanied by a decrease in SRF dependent gene expression and lethality at E11.5.

These mice exhibit several cardiac defects including abnormal cardiac looping and trabeculation, with disorganization of sarcomeres and Z discs. They also show a decrease in vascular SM cell recruitment to the dorsal aorta and disorganization of actin and

6

intermediate filaments in SM vasculature (Miano et al., 2004). Specific deletion of SRF in skeletal muscle causes severe skeletal muscle hypoplasia and perinatal lethality (Li et al., 2005b). These studies demonstrate the multifunctionality of SRF and its requirement for growth and differentiation of multiple organs, including brain and muscle.

Domain structure of MRTFs

Members of the Myocardin family of transcriptional co-activators include Myocardin,

MRTF-A, MRTF-B, and the MEF2 Activating SAP Transcriptional Regulator (MASTR), encoded by genes that span 93, 37, 50 and 5 kb on mouse chromosomes 11, 15, 16 and 7, respectively. Myocardin, MRTF-A, and MRTF-B are about 35% identical and share evolutionarily conserved domains required for SRF binding, homo- and hetero- dimerization, high-order chromatin organization, and transcriptional activation. However,

MRTF-A and -B differ from Myocardin and MASTR in their tissue distribution, domain structure, cellular localization, and responsiveness to stress signals. Hence, the amino acid identity between MRTF-A and -B is greater than that between other MRTF members

(Wang et al., 2002). Whereas myocardin expression is restricted to cardiac and smooth muscle cells and MASTR expression is limited to cardiac and skeletal muscle, MRTF-A and -B are expressed in a broad range of embryonic and adult tissues where SRF is expressed. Moreover, unlike MRTFs, whose cellular localization is regulated by actin binding, Myocardin and MASTR do not bind actin and are strictly localized to the nucleus (Creemers et al., 2006; Guettler et al., 2008).

All MRTF family members possess a conserved SAP (SAF-PIAS-Acinus) domain that interacts with matrix attachment regions to form active transcription domains

7

Figure 1.1. Domain Structure of the Myocardin Related Transcripton Factors (MRTFs). Shown are the RPEL motifs at the N-terminus, the basic (++) polyglutamine (Q) SAP domains, and the C-terminal transactivation domain, in addition to a MEF2 interacting domain unique for myocardin and MASTR.

8

and stimulate gene transcription. The SAP domain of Myocardin for instance mediates binding to histone acetyltransferase p300 and class II histone deacetylases to regulate the acetylation of nucleosomal histones surrounding SRF-binding sites in smooth muscle genes (Cao et al., 2005; McDonald et al., 2006). However, MRTFs can bind to various components of the Facilitating Chromatin Transcription (FACT) complex including

SPT16 and SSRP1 outside of the SAP domain to facilitate the transcriptional activity of

RNA polymerase II along nucleosomes (Kihara et al., 2008).

MRTFs associate with SRF via a short peptide sequence that includes a basic and a poly-glutamine (Q) rich region to form a powerful transcriptional complex. The Q-rich domain is also thought to mediate interactions with components of the transcriptional machinery, and its length correlates with the potency of the MRTF/SRF complex (Pipes et al., 2006; Wang et al., 2002). Binding of MRTFs to SRF adds a beta-strand to the

DNA binding beta-sheet region of SRF and bends the SRF/DNA complex to allow binding of MRTFs to DNA and stabilization of the tertiary MRTF/SRF/DNA complex

(Zaromytidou et al., 2006). However, although MRTFs seem to contact DNA flanking

CArG boxes, as yet no sequence-specific interactions have been identified.

MRTF-A and -B also contain three N-terminal RPEL motifs that bind monomeric

G-actin (Miralles et al., 2003; Mouilleron et al., 2008). In response to signals that promote actin polymerization, such as activation of the small GTPase Rho, MRTFs are released from G-actin, translocate to the nucleus, and activate transcription of SRF target genes including those encoding actin and other cytoskeletal components (Du et al., 2004;

Kuwahara et al., 2005; Pipes et al., 2006). MRTFs are thus direct mediators between actin dynamics and SRF transcriptional activity. The DNA binding function of the RPEL

9

Figure 2.2. Activation of MRTFs in response to extracellular signaling. In the cytoplasm, binding of MRTFs to monomeric actin inhibits their nuclear translocation. In response to extracellular signaling promoting actin polymerization, MRTFs translocate to the nucleus to activate SRF target genes.

10

motifs is not conserved in Myocardin and MASTR, which are constitutively active and irresponsive to cytoskeletal signaling.

Implication of MRTFs in multiple disease processes

MRTFs were first linked to disease development with the reported translocation of the human MRTF-A gene to chromosome 1 in acute megakaryocytic leukemia. This translocation creates a fusion protein with the human One-Twenty-Two (OTT) RNA binding protein resulting in constitutive activation of SRF dependent gene expression.

The OTT-MRTF fusion is constitutively nuclear, and is not regulated by actin signaling.

In addition to disrupting the MRTF dependent SRF activation, this fusion delays the activation of the MRTF independent TCF factors (Descot et al., 2008; Sawada et al.,

2008). Further studies with mouse MRTFs have shown their involvement in mediating

Rho dependent signaling to drive cell metastasis. Depletion of MRTFs or SRF in MDA-

MB-231 breast carcinoma and B16F2 melanoma cells reduces cell motility in culture and in xenografts and their rate of lung colonization from the bloodstream. Conversely, expression of activated MRTF-A increases lung colonization by the poorly metastatic

B16F0 cells (Medjkane et al., 2009).

In addition to their role in tumor development and metastasis, MRTFs have been recently implicated in the pathology of Alzheimer’s disease. Hypoxia was shown to stimulate the expression of SRF and Myocardin in human cerebral vascular SM cells and in animal models of Alzheimer’s disease. In addition, overexpression of SRF and

Myocardin in cerebral vascular SM cells disrupts the expression of low density lipoprotein receptor-related protein-1 impairing the clearance of amyloid beta-pepides

11

from the cerebral vasculature and controlling the progression of Alzheimer’s disease

(Bell et al., 2009). Moreover, disruption and activation of MRTF or SRF expression in mouse pial arteries disrupts arterial contractility and cerebral blood flow in response to brain activation in wild type and Alzheimer’s disease mouse models, implicating the

MRTF/SRF pathway in the progression of Alzheimer's dementia (Chow et al., 2007).

Myocardin and MASTR, coactivators of SRF and MEF2 in muscle cells

Myocardin, the founding member of the MRTF coactivators, was discovered using a bioinformatics-based screen designed to identify novel cardiac specific genes (Wang et al.,

2001). Myocardin is expressed as two alternatively spliced mRNAs (Creemers et al.,

2006; Wang et al., 2001). The two isoforms differ by the inclusion or exclusion of a unique peptide motif that binds to Myocyte Enhancer Factor (MEF2), another MADS box transcription factor, and are enriched in cardiac and smooth muscle tissue, respectively. MEF2 is involved in myogenesis and muscle function (Potthoff and Olson,

2007); hence, an interaction between Myocardin and MEF2 is thought to expand its regulatory potential and function in cardiac differentiation and function (Creemers et al.,

2006).

Myocardin is an essential regulator of smooth muscle gene regulation. In vitro, overexpression of Myocardin is sufficient to establish a contractile phenotype in smooth muscle cells in vitro (Long et al., 2008). In vivo, Myocardin knockout mice die embryonically by E10.5 due to disrupted smooth muscle differentiation (Li et al., 2003).

Moreover, conditional deletion of Myocardin in neural crest-derived smooth muscle cells

12

blocks the expression of smooth muscle restricted contractile proteins and results in persistent ductus arteriosis (Huang et al., 2008).

A role for Myocardin in cardiac development and pathology has been postulated based on studies performed in cardiomyocytes in vitro, using cardiomyocyte cultures, and in vivo, using Xenopus embryos. Myocardin is expressed in the embryonic mouse heart as early as E8.0 (Wang et al., 2001). Its expression is increased in the failing human heart compared to nonfailing heart samples (Torrado et al., 2003). Consistent with a role for Myocardin as a transducer of hypertrophic signals, forced expression of myocardin in cardiomyocytes is sufficient to induce cardiomyocyte hypertrophy and activation of a fetal gene program. Conversely, a dominant-negative mutant form of Myocardin, which retains the ability to associate with SRF but is defective in transcriptional activation, blocks cardiomyocyte hypertrophy induced by the hypertrophic agonist phenylephrine

(Xing et al., 2006). In Xenopus embryos, overexpression of Myocardin is sufficient to drive cardiac and smooth muscle differentiation markers in non-muscle cells; whereas knockdown of Myocardin impairs cardiac development and inhibits the expression of cardiac differentiation markers (Small et al., 2005). However, no cardiac function for

Myocardin has been reported in mammals yet.

MASTR, a recently identified SAP domain containing protein, functions as a coactivator of MEF2. MASTR and Myocardin share homologies in their SAP and MEF2 binding domains; however, unlike Myocardin, whose expression is cardiac and smooth muscle specific, MASTR is expressed in brain, cardiac and skeletal muscle (Creemers et al., 2006). Using knockdown and overexpression studies in Xenopus embryos, MASTR was shown to modulate the expression of skeletal muscle marker genes (Meadows et al.,

13

2008). However, its putative functions in cardiac and skeletal muscle development in mammals remain to be described.

Contribution of MRTF-A and -B to SRF signaling

Both MRTF-A and -B transcripts are detectable throughout the embryo at embryonic day

(E) 10.5. By E15.5, MRTF-A is expressed in a subset of neural mesenchymal cells, in skeletal muscle of the tongue, and in epithelial cells of the lung, kidney, bladder, colon and small intestines. Unlike MRTF-A, MRTF-B is expressed in both epithelial and mesenchymal cells of the lung, kidney, colon, small intestines and testis (Wang et al.,

2002).

The physical interaction between SRF and members of the MRTF family of coactivators has been well documented in vitro (Liu and Olson, 2006; Pipes et al., 2006), however the importance of these interactions in vivo remains to be understood. In vitro,

MRTF function has been studied in C2C12 skeletal muscle cells, which express both

MRTF-A and -B but not Myocardin. Expression of the dominant negative MRTF-B in

C2C12 myoblasts blocks their differentiation into myotubes and impairs the expression of SRF dependent differentiation genes, such as skeletal actin and heavy chain, implicating MRTFs in myogenic differentiation (Selvaraj and Prywes, 2003). In vivo, loss of function and dominant negative studies of the Drosophila MRTF (DMRTF) gene results in abnormalities in mesoderm migration, tracheal branching, and wing intervein development, which are reminiscent of SRF gain and loss of function mutations, suggesting an ancient partnership between MRTFs and SRF to direct cytoplasmic outgrowth and cell migration during development (Han et al., 2004). Moreover,

14

expression of a dominant negative MRTF mutant in skeletal muscle phenocopies the phenotype observed with skeletal muscle deletion of SRF, suggesting that the

MRTF/SRF partnership is conserved in mammals and required for skeletal muscle growth and maturation (Li et al., 2005b).

However, mice lacking any single member of the MRTF family fail to phenocopy

SRF loss of function mutations. MRTF-A knockout mice have defects in mammary myoepithelial cell differentiation but appear otherwise normal (Li et al., 2006; Oh et al.,

2005); whereas global deletion of MRTF-B causes defects in the patterning of the branchial arch arteries that results postgastrulation embryonic lethality (Li et al., 2005a;

Oh et al., 2005). As stated earlier, Myocardin knockout mice die embryonically due to disrupted smooth muscle differentiation (Li et al., 2003). These studies suggest that in vivo SRF activity relies on transcriptional cofactors outside of the MRTF family or that functional redundancy among MRTF members have obscured in vivo analyses with single gene mutations. In fact, in vitro studies demonstrate the redundancy between

MRTF-A and -B in multiple cell lines. For instance, deletion of both MRTF-A and -B is required to inhibit the activation of SRF by Rho signaling in HeLa or NIH 3T3 cells (Cen et al., 2003). However, MRTF-A and -B are coexpressed in mammary myoepithelial cells and in vascular SM cells of the aorta and bronchial arteries, suggesting either that

MRTFs have distinct functions in these tissues or that different tissues are differentially sensitive to the levels of MRTF expression.

Putative MRTF functions in brain and muscle

15

Numerous cellular processes, such as cell migration, neurite outgrowth, and sarcomere assembly rely on precisely coordinated communication between the cytoskeleton and the genome. Because of its crucial role in maintaining cell shape and regulating nuclear gene expression, the dynamics of the actin cytoskeleton are subject to tight regulation to support normal cellular functions (Stern et al., 2009). SRF plays an essential role in controlling cytoskeletal assembly by regulating the expression of cytoskeletal and sarcomeric components including actins, , tropomyosins, vinculin and others

(Posern et al., 2004; Sun et al., 2006). Mechanistically, SRF senses the state of actin polymerization to modulate the expression of cytoskeletal components and thereby to regulate neuronal and muscle development and function (Kuwahara et al., 2005; Miralles et al., 2003; Posern and Treisman, 2006). The functions of MRTFs in these processes remain to be determined.

In the brain, MRTF-A and -B are coexpressed in the forebrain, particularly in the hippocampus and cerebral cortex (Alberti et al., 2005; Shiota et al., 2006). In the heart,

MRTF-A and -B are coexpressed with Myocardin and MASTR. As stated earlier, generation of mouse models with brain and cardiac specific SRF mutant alleles elucidated the role of SRF in the development and maintenance of neuronal and cardiac function (Alberti et al., 2005; Knoll et al., 2006; Niu et al., 2005; Parlakian et al., 2005;

Parlakian et al., 2004); however, the MRTF knockout mice do not exhibit obvious neuronal or cardiac abnormalities (Li et al., 2005a; Li et al., 2006; Li et al., 2003; Oh et al., 2005).

Based on all of the above, I set out to generate mice that lack both MRTF-A and -

B as a tool to elucidate the putative redundant roles of these proteins and to examine the

16

mechanisms that distinguish the functions of different MRTF members, when coexpressed in the same tissue. I chose to examine these roles in the brain, where

Myocardin is not expressed, and where cytoskeletal dynamics and SRF activity are indispensable for development and plasticity. I also chose to examine MRTF double knockout mice in the heart, where Myocardin is also expressed, to address the potential requirement for a threshold of MRTF activity for cardiac development and possible differential functions of MRTFs in the heart. Moreover, with the identification of

MASTR as a novel MRTF member, I also examined the functions of MASTR in muscle and brain.

Therefore, to determine the functions of MRTFs during neuronal and muscle development, my specific aims were:

1. To determine the in vivo function of MRTF-A and -B during brain development.

2. To determine the in vivo function of MRTF-A and -B in cardiac development and

function.

3. To determine the function of MASTR in muscle and brain development.

17

Chapter II

MRTFs Regulate the Cdk5/Pctaire-1 Kinase cascade to Control Neurite Outgrowth and

Neuronal Migration during Brain

Development

18

ABSTRACT

The actin cytoskeleton is a dynamic structure required for diverse cellular processes including cell migration, cell adhesion, and neurite outgrowth. The modulation of cytoskeletal structure and function in response to extrinsic and intrinsic cellular cues depends on communication between the cell membrane and the nucleus. Members of the

Myocardin family of transcriptional coactivators provide transcriptional potency for serum response factor (SRF), a regulator of genes involved in cell proliferation, migration, and cytoskeletal dynamics. Here, we report that conditional deletion of the

Myocardin-related transcription factor (MRTF) -A and -B in the mouse brain disrupts neuronal migration and neurite outgrowth. Inappropriate neurite outgrowth in these double knockout mice causes aberrant development of multiple brain structures including the hippocampus, the cerebral cortex, and the subventricular zone. We show that MRTFs organize the actin cytoskeleton by regulating the expression and the activity of the actin severing proteins Gelsolin and Cofilin. Finally, we identify Pctaire-1 as a novel SRF target gene and a regulator of neurite outgrowth in primary neuronal cultures. Our study uncovers a novel protein kinase cascade whose core component Pctaire-1 is activated by the MRTF/SRF pathway and whose function is to regulate actin dynamics, neurite outgrowth and neuronal migration during neurogenesis.

19

INTRODUCTION

Precisely controlled neuronal migration and neuronal connectivity establishes normal brain architecture and maintains neurological and cognitive functions (Ayala et al., 2007;

Lin et al., 2009). Aberrant neuronal migration or connectivity causes multiple neurodevelopmental and neurodegenerative disorders including mental retardation, schizophrenia, Rett’s syndrome, Alzheimer’s disease, and Parkinson’s disease. Although these disorders differ in their onsets and pathologies, they share common etiologies including the absence of functional neurons in discrete regions of the brain, decreased neurite arborization, and weak synapse connections. Elucidating the molecular pathways controlling neuronal survival, migration, and connectivity could identify novel therapeutic targets for enhancing neuronal function in these brain disorders (Benitez-King et al., 2004; Lin et al., 2009; Spalice et al., 2009; Verrotti et al., 2009).

Neurons originate in the ventricular zones of the brain and undergo stereotypical migration to populate specific brain structures. Neuronal migration depends on neurite extension and outgrowth, during which neurons dynamically generate and retract growth cones in search of extracellular guidance cues. In response to an attractant, a growth cone will extend to form a leading neurite and initiate neuronal migration. The nucleus then translocates into the leading neurite while the trailing process retracts (Ayala et al.,

2007; Lambert de Rouvroit and Goffinet, 2001). Extracellular guidance molecules including netrins, slits, and semaphorins; the cell-adhesion complexes cadherins, laminins, and reelins; and the neurotrophic factors BDNF and NT4 instruct neurons to migrate directionally to specific sites in the brain during neurogenesis (Lin et al., 2009; Park et al.,

2002). The expression of these guidance factors is sustained in the adult brain and

20

modified following injury to regulate neurite outgrowth and, in turn, the reorganization of neuronal connections in response to activity or injury (Ayala et al., 2007; Rossi et al.,

2007).

Dynamic changes in the actin cytoskeleton provide the mechanical force for neurite outgrowth and migration. Actin polymerization drives neurite extension and requires a pool of monomeric G-actin to extend filamentous F-actin fibers toward the leading edge of the outgrowth. Guidance molecules orchestrate directional cell movement by regulating the transcription of structural cytoskeletal components, including actin, and the activity of cytoskeletal regulatory proteins, such as the F-actin severing proteins Gelsolin and Cofilin (Cunningham et al., 1991; Furnish et al., 2001; Lin et al., 2009; Meberg et al., 2000; Meberg, 2000; Meberg and Bamburg, 2000; Park et al.,

2002; Stern et al., 2009; Tanaka et al., 2008). The intracellular pool of available G-actin governs the rate of actin polymerization and is regulated at both the transcriptional and posttranscriptional levels by extracellular signals. However, the intracellular regulatory cascades that sense G-actin levels and provide the appropriate intracellular response to guidance cues remain poorly understood.

Members of the myocardin family of transcription factors modulate cytoskeletal dynamics by sensing actin polymerization and conferring transcriptional activity to

Serum Response Factor (SRF). Unlike Myocardin, whose expression is restricted to cardiac and smooth muscle (Oh et al., 2005; Wang et al., 2002), the expression Mrtf-A and -B is enriched in the forebrain, particularly in the hippocampus and cerebral cortex

(Alberti et al., 2005; Shiota et al., 2006). Studies in muscle cells show that Mrtf-A and -

B translocate between the cytoplasm and the nucleus in response to extracellular

21

signaling. Mrtfs bind monomeric G-actin in the cytoplasm via their three N-terminal

RPEL motifs. In response to signals promoting actin polymerization, Mrtfs are released from G-actin, translocate to the nucleus, and form powerful transcriptional complexes with Serum Response Factor (SRF). Within the nucleus Mrtfs co-activate a number of

SRF target genes including those encoding actin and other cytoskeletal components

(Kuwahara et al., 2005; Miralles et al., 2003; Pipes et al., 2006).

Mouse embryos homozygous for an SRF null mutation fail to generate mesoderm and die at gastrulation (Arsenian et al., 1998), however tissue specific deletion of SRF has revealed multiple roles for this transcription factor in brain, muscle, heart, and liver development (Charvet et al., 2006; Latasa et al., 2007; Miano et al., 2004; Niu et al.,

2005; Parlakian et al., 2005). Conditional deletion of SRF in the brain impairs neurite outgrowth, axon guidance, directional migration of neurons from the subventricular zone to the olfactory bulb, and learning and memory (Alberti et al., 2005; Etkin et al., 2006;

Knoll et al., 2006; Ramanan et al., 2005). However, mice lacking any single member of the Myocardin family fail to phenocopy SRF loss-of-function mutations. MRTF-A knockout mice have defects in mammary myoepithelial cell differentiation but appear otherwise normal (Li et al., 2006; Oh et al., 2005) whereas global deletion of MRTF-B causes defects in the patterning of the branchial arch arteries and postgrastrulation embryonic lethality (Li et al., 2005a; Oh et al., 2005). These studies suggest that in vivo

SRF activity relies on transcriptional cofactors outside of the Myocardin family or that functional redundancy among MRTF members have obscured in vivo analyses with single gene mutations.

22

Here, we show that a single allele of either MRTF-A or -B is sufficient to support normal brain development, however brain-specific deletion of MRTF-B in MRTF-A null mice causes lethality between postnatal day (P) 16 and 21. Mice lacking both MRTF-A and -B in the brain show morphological abnormalities in the hippocampus, cerebral cortex, and subventricular zone (SVZ) that phenocopy the defects reported for brain- specific deletion of SRF (Alberti et al., 2005; Knoll et al., 2006). The morphological defects in MRTF-A/B mutant mice are accompanied by aberrant neuronal migration, impaired neurite outgrowth, and decreased expression and activity of actin and the actin severing proteins Gelsolin and Cofilin. We also identify the gene encoding Pctaire-1 kinase as a novel target of the MRTF/SRF pathway in the brain. Pctaire-1 links

MRTF/SRF signaling to the Cdk5 pathway, a known regulator of actin dynamics that promotes neuronal migration and neurite outgrowth. We conclude that MRTF-A and -B are key molecular sensors in a tightly controlled regulatory feedback loop that links extracellular signaling and cytoskeletal activity with SRF transcriptional regulation to modulate actin dynamics during neurogenesis in vivo.

RESULTS

Generation of mice lacking MRTF-A and MRTF-B in the brain

To explore the potential functions of MRTFs in postnatal development, we generated a conditional null MRTF-B allele by introducing loxP sites within introns 7 and 8 of the mouse MRTF-B locus by homologous recombination in embryonic stem cells (Figure

2.1) (Collaboration with Yuri Kim). Deletion of exon 8, which encodes the basic domain and part of the poly-glutamine domain of MRTF-B, eliminates binding to SRF, thereby

23

Figure 2.1. Generation of MRTF brain double knockout (bdKO) mice. (A) Strategy to generate a conditional MRTF-B allele. Protein, corresponding exonic structure, and targeted alleles are shown. (B) Southern blot analysis confirming targeting of ES cells. The corresponding wild type and targeted bands are indicated for the 5’ and 3’ probes. (C) Genotyping of MRTF-BF and MRTF-BKO mice by genomic PCR. (D) MRTF bdKO mouse (left) and control MRTF-A-/+; MRTF-BF/F;GFAP Cre mouse (right) are shown at P14. (E) Average weight of control and bdKO mice at P14. The MRTF bdKO mice are significantly smaller than their control littermates. (F) MRTF takeout in MRTF bdKO brains as assessed by quantitative RT-PCR.

24

allowing analysis of the influence of MRTF on SRF activity in vivo. The MRTF-Bneo-loxP mice were bred to Flpe transgenic mice to obtain the “floxed” MRTF-B allele, hereafter referred to as MRTF-BF. Correct targeting and germline transmission were confirmed by

Southern blot and PCR of genomic DNA (Figure 2.1). Specific deletion of MRTF-B in the brain was achieved by crossing MRTF-BF/F mice to transgenic mice expressing Cre recombinase under the control of the human GFAP promoter, which directs expression in radial glial cells as early as E12.5. Radial glial cells give rise to glial cells, to the majority of neurons in the cerebral cortex, hippocampus, and cerebellum, and to adult

SVZ stem cells that produce neurons throughout adult life (Zhuo et al., 2001). MRTF-

BF/F;GFAP-Cre mice were phenotypically normal and displayed no obvious phenotype.

Given the degree of homology between MRTF-A and -B and the expression of both MRTFs in the developing and adult nervous system (Liu and Olson, 2006; Pipes et al., 2006; Shiota et al., 2006), we speculated that MRTF-A could compensate for MRTF function in the brains of MRTF-B null mice. To test this hypothesis, we crossed our

MRTF-B conditional mice to mice harboring an MRTF-A null (MRTF-A-/-) allele (Li et al., 2006) to generate mice lacking both MRTF-A and -B in the brain. We refer to the

MRTF-A-/-; MRTF-BF/F; GFAP-Cre mice as MRTF brain double knockout (MRTF bdKO) mice.

MRTF bdKO mice were born at Mendelian ratios, but showed a significant reduction in body size and weight and diminished locomotor activity by P14 (Figure 2.1).

All MRTF bdKO mice died between P16-P21. Quantitative real-time PCR, using primers located within the deleted exons of either MRTF-A and MRTF-B, confirm significant MRTF takeout in MRTF bdKO brains compared with control brains (Figure

25

2.1). Interestingly, MRTF-A-/+;MRTF-BF/F; GFAP-Cre and MRTF-A-/-;MRTF-BF/+;

GFAP-Cre mice were phenotypically normal, suggesting that a single copy of MRTF-A or -B is sufficient to rescue the phenotype.

MRTF bdKO mice have severe brain abnormalities

Histological analyses of brain sections from MRTF bdKO mice at P16 revealed several morphological abnormalities in the hippocampus, the corpus callosum, the anterior commissures, and the striatum that phenocopy defects reported in mice with brain- specific deletion of SRF (Alberti et al., 2005). Hematoxylin and eosin (H&E) staining of sagittal brain sections from MRTF bdKO mice showed a reduction in the size of the striatum and an expansion of the SVZ (Figure 2.2). H&E staining of coronal sections from MRTF bdKO mice revealed an absence of both the corpus callosum and the anterior commissures that is accompanied by a compressed hippocampus with a deformed dentate gyrus (Figure 2.2). Nissl staining of hippocampal neurons identified a poorly defined pyramidal cell layer in MRTF bdKO mice, a phenotype that is confirmed by Golgi staining (Figure 2.2). Golgi staining also showed a dramatic decrease in the number of hippocampal neuronal projections compared to control neurons (Figure 2.1).

Defective neuronal migration along the rostral migratory stream

The accumulation of cells at the SVZ in MRTF bdKO brains could be observed as early as P1. Normally, newborn neurons originating from the SVZ migrate out tangentially along the rostral migratory stream (RMS) towards the olfactory bulbs where they differentiate into interneurons (Figure 2.4). We speculated that the phenotype we

26

Figure 2.2. Neuroanatomical defects in MRTF bdKO mice. (A-F) Hematoxylin and eosin (H&E)-stained brain sections of control (A,C,E) and MRTF bdKO brains (B,D,F) at P16. bdKO mice show defects in the striatum (St), subventricular zone (SVZ), corpus callosum (CC), anterior commissures (AC), hippocampus and dentate gyrus (DG) (Scale bar: 500mm). (G,J) Defective hippocampal neuronanatomy of the MRTF bdKO mice. Nissl staining reveals a loosely defined pyramidal cell layer (PL) of the hippocampi of MRTF bdKO mice (H) compared to their control littermates (G). Golgi staining reveals defects in the arrangement of the pyramidal neurons and diminished neuronal projections within the hippocampi in the MRTF bdKO brains (J) compared to control brain sections (I) (Scale bar: 100mm).

27

observed in MRTF bdKO mice at the SVZ was due to a neuronal migration defect. This hypothesis is supported by the fact that MRTF-A and -B null mouse embryonic fibroblasts (MEFs) fail to arrange their cytoskeleton and migrate to the wound site in an in vitro scratch assay (Figure 2.3). Alternatively, SVZ expansion could result from excessive proliferation of neuronal precursors at the SVZ. To distinguish between these possibilities, we used BrdU labeling to monitor cellular proliferation and migration from the SVZ towards the olfactory bulb. BrdU pulse labeling for one hour at E12.5 and

E15.5 did not reveal a difference in the rate of cell proliferation at the SVZ (Figure 2.4)

(data not shown).

We then chased the E15.5 BrdU labeled neurons by analyzing their number and position in P7 mice. In control mice, the majority of BrdU positive cells had migrated away from the SVZ by P7; whereas MRTF bdKO mice showed a significant accumulation of BrdU positive cells at the SVZ (Figure 2.4). As a consequence of the defective neuronal migration along the RMS, the number of neurons at the olfactory bulbs in the MRTF bdKO brains was significantly diminished compared to control brains

(Figure 2.4). Moreover, terminal deoxynucleotidyl biotin-dUTP nick end labeling (TUNEL) staining revealed increased apoptosis within the striatum of MRTF bdKO mice compared to their control littermates (Figure 2.4). These results demonstrate that the expanded SVZ we observed in MRTF bdKO mice is due to defective neuronal migration and further support the hypothesis that MRTFs regulate cyotskeletal dynamics in the developing nervous system.

28

29

Figure 2.3. Deletion of MRTF-A and -B in mouse embryonic fibroblasts (MEFs). (A) Defective migration of MEFs following MRTF deletion. Mouse embryonic fibroblasts derived from MRTF-A-/+;MRTF-BF/F or MRTF-A-/-;MRTF-BF/F mouse embryos were infected with either GFP-Cre-expressing or control GFP-expressing adenovirus and subjected to wound healing assay. (B) Disruption of actin dynamics following MRTF deletion in MEFs. MEFs derived from MRTF-A-/+;MRTF-BF/F or MRTF-A-/-;MRTF-BF/F mouse embryos were infected with either GFP-Cre-expressing or control GFP-expressing adenovirus and stained with phalloidin. Note the disorganization of actin filaments and presence of cortical actin staining upon MRTF deletion.

30

Figure 2.4. Defective neuronal cell migration in MRTF bdKO mice. (A) Schematic diagram shows the migration of neurons from the SVZ along the RMS to reach the olfactory bulbs (OB). This process is disrupted in MRTF bdKO mice. (B) Impaired migration of neuronal cells from the SVZ along the RMS in bdKO mice. BrdU labeling of sagittal brain sections reveals the accumulation of BrdU positive cells in the SVZ of bdKO brains. (C) Counts of neurons at the olfactory bulb are significantly reduced in bdKO mice (p<0.0001) (40 fields total from 4 brains) (D) H&E-stained brain sections at the level of the olfactory bulb show a decrease in the number of interneurons in bdKO mice compared to their control littermates. (E,F) One hour BrdU pulse labeling of proliferating neuronal precursors at the SVZ shows comparable levels of neuronal proliferation between control (E) and bdKO (F) brain sections. (G,H) TUNEL staining of control (G) and MRTF bdKO (H) brain sections shows increased apoptosis in the striatum of MRTF bdKO mice compared to control mice (Scale bar: 200mm). 31

Defective neurite outgrowth in MRTF bdKO neurons

In addition to defects in neuronal cell number, organization, and position, we also observed by Golgi staining that the number of projections from hippocampal neurons was dramatically reduced in MRTF bdKO mice at P16 (Figure 2.2). Considering the role of the MRTF/SRF pathway in regulating cytoskeletal genes and the implication that SRF participates in neurite outgrowth (Knoll et al., 2006), we wanted to further investigate the role of MRTFs in directing neuronal projections outside the hippocampus. We performed anti-MAP-2 antibody staining to examine the organization of neuronal extension in vivo. Interestingly, MRTF bdKO mice show less MAP-2 in the striatum and cortical regions of the brain compared to control animals (Figure 2.5). Defective neuronal projections in the cortex was also confirmed by Golgi staining (Figure 2.5).

To confirm that the decrease in MAP-2 staining is indeed indicative of a defect in neurite extension, we performed an in vitro neurite outgrowth assay in which hippocampal and cortical neurons were cultured from MRTF-A-/-; MRTF-BF/F P1-P3 mice and infected with GFP-Cre-expressing adenovirus. Neurons from MRTF-A-/+;

MRTF-BF/F mice infected with GFP-Cre-expressing adenovirus and neurons from

MRTF-A-/-; MRTF-BF/F mice infected with GFP-expressing adenovirus were used as controls. 80% of the GFP positive cells in control cultures formed readily identifiable neurites, however only about 15% of the MRTF bdKO neurons grew neurites over the two-week culture period (Figure 2.5). The same assay was used to examine neurite outgrowth in primary cultures of MRTF bdKO neurons over a one-week culture. In this assay, 75% of neurons in control cultures formed neurites, compared to less than 20% in

32

Figure 2.5. Decreased neuronal projections in MRTF bdKO mice. (A-D) Reduced MAP-2 staining in the striatum (A,B), and cortical (C,D) regions of MRTF bdKO mice at P16 (Scale bar: 50mm). (E,F) Golgi staining reveals defects in the arrangement of cortical neurons and decreased neuronal projections in the cortical regions of MRTF bdKO brains (F) compared to the control brain sections (E) (Scale bar: 100mm). (G-I) Hippocampal neurons derived from MRTF-A-/+;MRTF-BF/F or MRTF-A-/-;MRTF-BF/F mice were infected with either GFP-Cre-expressing (H) or control GFP-expressing (G) adenovirus. Shown are the GFP and anti-MAP-2 antibody stainings from two-week neuron cultures (Scale bar: 10mm). (I) Percent neurite outgrowth represents the percent of GFP-positive cells that show neurite outgrowth. Significant decrease in the percent of neurite outgrowth in MRTF-A-/-;MRTF-BF/F Cre-infected neurons compared to control neurons (45 fields from 3 different experiments) (p<0.03). (J-L) Neurite outgrowth in hippocampal neurons derived from MRTF bdKO or control MRTF-A-/+; MRTF-BF/F mouse pups. Shown is anti-MAP-2 antibody staining from one-week neuron cultures (Scale bar: 10mm). (L) Significant decrease in the percent of neurite outgrowth in the bdKO culture compared to the control culture (30 fields from 3 different experiments) (p<0.0001).

33

the MRTF bdKO neuronal cultures (Figure 2.5). Taken together, these studies show that

MRTFs induce neurite outgrowth in multiple regions of the forebrain.

Pctaire-1 is a novel MRTF/SRF target gene

In order to explore the molecular basis for the abnormalities in neurite outgrowth and migration in MRTF bdKO mice, we performed microarray analysis on cortical samples from control and MRTF bdKO brains at Po. As expected, the expression of numerous known SRF target genes was diminished in the MRTF bdKO mice. We were able to confirm the downregulation of beta-actin and the actin binding protein gelsolin in the hippocampal and cortical regions of the MRTF bdKO brains by quantitative real-time

PCR (Figure 2.6).

In addition to these known SRF target genes we were interested in a small set of genes that were downregulated in the microarray, were not known SRF target genes, and that function during neuronal development. Among the genes fitting these criteria, the protein kinase Pctaire-1 was one of the most downregulated genes in the microarray.

Gene expression analysis by quantitative real-time PCR confirmed downregulation of

Pctaire-1 in the cortical and hippocampal regions of MRTF bdKO mice (Figure 2.7).

Interestingly, expression of Pctaire-2, a closely related Pctaire family member, was unaltered in MRTFs bdKO mice (Figure 2.7). Furthermore, anti-Pctaire-1 staining revealed decreased expression of Pctaire-1 in the pyramidal cells of the hippocampus and in the cortex of MRTF bdKO mice compared to control mice (Figure 2.7).

We analyzed the DNA sequence upstream of the Pctaire-1 gene and identified a conserved CArG box located 400 base pairs upstream of the transcriptional start site. Gel

34

Figure 2.6. Aberrant expression of actin and Gelsolin in MRTF bdKO brains. Quantitative RT-PCR analysis from hippocampal (grey) and cortical (black) regions of MRTF bdKO brains. Note the decreased expression of beta-actin and Gelsolin (Gsn); whereas expression of Cofilin (Cf1) at the mRNA level is unchanged. Relative expression was normalized to GAPDH levels and to the corresponding transcript levels in control brains.

35

Figure 2.7. Identification of Pctaire-1 as a novel MRTF/SRF target gene. (A) Decreased expression of Pctaire-1 transcripts in MRTF bdKO brains. Quantitative RT- PCR from hippocampal and cortical regions of MRTF bdKO brains. (B) Anti-Pctaire-1 antibody staining revealed decreased levels of the protein in the hippocampal and cortical regions of MRTF bdKO mice compared to control mice (Scale bar: 50mm). (C) Gel mobility shift assay shows binding of GST-SRF to the Pctaire-1 CArG sequence. (D) Responsiveness of the Pctaire-1 gene expression to MRTF/SRF signaling. Increased amounts of the SRF were able to activate a Pctaire-1-luciferase reporter construct but not an empty luciferase contruct. Mutation of the predicted CarG box blocks this activation (E) Defective neurite outgrowth in Pctaire-1 knockdown neurons. Anti-MAP-2 antibody staining from one-week neuron cultures disrupts the growth of neurites in the Pctaire-1 siRNA but not in control siRNA transfected neurons (Scale bar: 5mm). 36

mobility shift assays showed that this CArG box forms a DNA-protein complex with recombinant GST-SRF protein (Figure 2.7). Protein binding was disrupted following mutation of the CArG box and was competed by the unlabeled wild-type probe, but not by the mutated probe. We then generated a luciferase reporter construct containing the

500bp region of Pctaire-1 upstream of the transcriptional start site that includes the putative CArG box. In this assay, we used a constitutively active nuclear localized

Myocardin to activate MRTF/SRF signaling. Myocardin and SRF activated this reporter in a dose dependent manner and this activation is blunted in a reporter containing a mutated CArG box (Figure 2.7). Pctaire-1 is thus a bona fide SRF target gene whose expression is MRTF dependent in the developing forebrain.

These results suggested to us that Pctaire-1 could be an essential effector of the

SRF/MRTF pathway during neurite outgrowth. To further study the role of Pctaire-1 on neuronal cell morphology and neurite outgrowth, we performed knockdown experiments in which we transfected primary neuron cultures with three different siRNAs for Pctaire-

1 or a control siRNA. Pctaire-1 knockdown disrupted neurite outgrowth in our primary culture assay and phenocopies MRTF-A and -B null neuronal cultures (Figure 2.7).

Regulation of actin dynamics by MRTFs.

Considering the reported interaction between Pctaire-1 and Cdk5, we wondered whether

MRTFs regulate Cdk5 activity in the brain. To assess Cdk5 activity we performed immunohistochemistry against the activated (phosphorylated) form of Cdk5 and observed reduced ph-Cdk5 accumulation in the hippocampal and cortical regions of MRTF bdKO mice compared to control littermates (Figure 2.8) (data not shown). To further confirm

37

Figure 2.8. Regulation of actin dynamics by the MRTF/SRF pathway. (A-F) Dysregulation of the phosphorylation of Cdk5, LimK, and Cofilin in MRTF bdKO brains. Anti-ph-Cdk5 (A,B), anti-ph-LimK (C,D), and anti-ph-Cofilin (E,F) staining revealed decreased phosphorylation of Cdk5 and increased phosphorylation of LimK and Cofilin in the brains of MRTF bdKO mice (B, D, and F) compared to control mice (A, C, and E). (G,H) Phalloidin staining revealed decreased levels of F-actin in the hippocampal regions of MRTF bdKO mice (H) compared to control mice (G). (I) Diminished kinase activity of Cdk5 upon MRTF deletion. Kinase assays show 75% decrease in the kinase activity of Cdk5 immunoprecipitated from the brains of MRTF bdKO mice compared to control mice (J) Western blot analysis of phospho-Cofilin and total Cofilin in the cerebellar cortex of control and MRTF bdKO brain samples. GAPDH was used as a loading control (Scale bar: 50mm).

38

abrogated activity of Cdk5 in the MRTF bdKO brains, we performed a Cdk5 kinase assay with immunoprecipitated Cdk5 from the cortical regions of either control or bdKO mice and used recombinant histone H1 protein as substrate. Indeed, the kinase activity of

Cdk5 was significantly reduced in the MRTF bdKO brains compared to control brains

(Figure 2.8).

Cdk5 regulates neuronal migration and neurite outgrowth by modulating the activity of a kinase cascade that targets actin polymerization. At the base of this cascade is the actin depolymerase Cofilin whose activity is diminished by phosphorylation.

Cofilin is a substrate for Lim Kinase (LimK) that is in turn a substrate for the kinase Pak1.

Thus activated Pak1 enhances actin polymerization by reducing Cofilin activity. Cdk5 on the other hand inactivates Pak1 via phosphorylation and promotes actin depolymerization. We speculated that the Pak1-Limk-Cofilin cascade is a target of Cdk5 downstream of MRTFs. Indeed, the cortical and hippocampal regions of the MRTF bdKO mice showed greater accumulation of ph-LimK and ph-Cofilin compared to control brains (Figure 2.8) (data not shown). Furthermore, immunoblotting for ph-

Cofilin and total Cofilin showed a significant increase in ph-Cofilin in the brains of

MRTF bdKO mice as compared to controls without a significant change in total levels of

Cofilin protein (Figure 2.8). These results define a mechanistic pathway in which

MRTFs induce Cdk5 activity and Cdk5 in turn inactivates the Pak1-Limk cascade to promote Cofilin actin depolymerase activity and neuronal outgrowth.

To verify that dysregulation of the two major actin severing proteins, Cofilin as well as Gelsolin abrogates actin dynamics, we evaluated the levels of actin polymerization by filamentous F-actin staining (phalloidin). Phalloidin staining of

39

MRTF-A and -B null MEFs showed actin cytoskeleton disruptions and lack cortical accumulation of actin filaments (Figure 2.3). The levels of F-actin were also markedly decreased in the cortical and hippocampal regions of MRTF bdKO mice compared to their control littermates (Figure 2.8). MRTFs are thus essential regulators of actin dynamics.

DISCUSSION

The results of this study reveal obligate roles for MRTF-A and -B as redundant regulators of neurite outgrowth and neuronal migration during brain development in vivo. The abnormalities in brain development resulting from the combined deletion of MRTF-A and -B reflect disorganization of the actin cytoskeleton and impairment of actin dynamics.

MRTFs couple the Rho and Cdk5 signaling pathways to actin treadmilling and SRF- dependent transcription to regulate neuronal development, as schematized in Figure 2.9.

Upstream signals that activate Rho signaling liberate MRTFs from monomeric G-actin, allowing MRTFs to translocate to the nucleus and enhance SRF activity. Targets of the

MRTF/SRF signaling pathway include genes encoding actin and other cytoskeletal components as well as Pctaire-1, a negative regulator of actin polymerization. MRTFs also promote the kinase activity of a second negative regulator of actin polymerization,

Cdk-5, likely at the post-transcriptional level. Thus, MRTFs function as nodal regulators that couple two downstream signaling pathways that modulate cytoskeletal dynamics, thereby fulfilling a cytoskeletal-transcriptional circuit that governs neurite outgrowth and neuronal development.

40

Figure 2.9. Proposed model for the regulation of neuronal migration and neurite outgrowth by the MRTF/SRF pathway. MRTFs are essential mediators of SRF signaling in the mouse brain and key regulators of the dynamics of the actin cytoskeleton. Together with SRF, MRTFs regulate the expression of beta-actin and other actin binding proteins (ABPs) such as Gelsolin. In addition, activation of the MRTF/SRF signaling regulates the expression of Pctaire-1, which is involved in the regulation of neurite outgrowth. Under normal conditions, the MRTF/SRF pathway regulates the activity of Cdk5, which phosphorylates and inactivates Pak1. Decreased Pak1 activity results in diminished LimK activity towards phosphorylation of Cofilin resulting in increased actin severing. Genetic ablation of MRTFs in the mouse brain disrupts actin dynamics by disrupting the expression and/or activity of actin and the actin severing proteins Gelsolin and Cofilin, resulting in defective neuronal migration and neurite outgrowth.

41

Functions of MRTFs in vivo

This study is the first to reveal to the importance of the MRTF/SRF partnership during brain development in vivo. Physical interaction between MRTFs and SRF and potentiation of the transcriptional activity of SRF by MRTFs have been well documented in vitro (Liu and Olson, 2006; Pipes et al., 2006). However, the phenotypes of knockout mice lacking Myocardin, MRTF-A or -B have not been comparable to those of SRF knockout mice. The remarkable similarities between the neuronal defects resulting from deletion of both MRTF-A and -B and those reported for brain-specific SRF knockout mice (Alberti et al., 2005; Knoll et al., 2006) strongly suggest that the actions of SRF in the brain rely completely on its association with MRTFs and argue against the involvement of other transcriptional cofactors in mediating SRF signaling during brain development. Neuronal defects were only observed upon deletion of all four MRTF alleles in the brain; a single copy of either MRTF-A or -B is sufficient to support normal brain development. The redundancy of MRTF functions in the brain contrasts with the distinct phenotypes observed in MRTF-A and MRTF-B single knockout mice, which show abnormalities in mammary myoepithelial cells and the branchial arch arteries, respectively (Li et al., 2005a; Li et al., 2006; Oh et al., 2005).

MRTFs as regulators of actin cytoskeleton dynamics

Precise control of the neuronal actin cytoskeleton in response to intrinsic and extrinsic cues governs multiple events during brain development such as neurite outgrowth and neuronal migration. Such control could be exerted by direct regulation of the expression of G-actin and by regulation of the activity of actin-binding proteins, which influence

42

actin dynamics. The assembly of G-actin into actin filaments is tightly regulated and crucial for maintaining the structure of the cytoskeleton and nuclear gene expression. In a recent report, Stern et al. made use of actin mutants that either favor or inhibit F-actin assembly to show that actin filaments can induce neurite outgrowth in neuronal cultures and that regulation of neurite length by actin filaments is mediated by the activity of SRF

(Stern et al., 2009). Our finding that deletion of MRTF-A and -B results in diminished assembly of actin filaments accounts for the defective neuronal migration and neurite outgrowth in the brains of MRTF bdKO mice.

Genetic ablation of MRTF-A and -B also results in decreased expression and activity of the actin-severing proteins Gelsolin and Cofilin at the transcriptional and posttranslational levels, respectively. Gelsolin and Cofilin are the two major actin- severing proteins, which mediate depolymerization and recycling of monomeric actin from actin filaments. Both proteins are enriched at growth cones and required for extension of neuronal processes and neurite outgrowth (Furnish et al., 2001; Meberg,

2000; Meberg and Bamburg, 2000). Overexpression of Gelsolin enhances cell motility

(Cunningham et al., 1991) and increases the length and motility of neurites (Furnish et al.,

2001). Genetic deletion of Gelsolin in fibroblasts results in disruption of the actin cytoskeleton and slower migration in vitro (Witke et al., 1995). Unlike Gelsolin, which does not require phosphorylation for activity, the actin-severing activity of Cofilin is regulated through phosphorylation by Lim , which function as downstream effectors of Rho GTPases (Abe et al., 1996; Kuhn et al., 2000; Sarmiere and Bamburg,

2004). Cofilin is an essential regulator of the actin cytoskeleton, critical for cell migration (Bellenchi et al., 2007; Gurniak et al., 2005) and neurite outgrowth (Meberg,

43

2000; Zhang et al., 2006). The actin severing activity of Gelsolin and Cofilin is required to maintain a high concentration of G-actin for continued polymerization at the barbed end of the actin filaments during neuronal migration and neurite outgrowth (Dent and

Gertler, 2003; Furnish et al., 2001; Meberg, 2000; Meberg and Bamburg, 2000).

Aberrant actin-severing could thus explain the defective organization of the actin cytoskeleton in the brains MRTF bdKO mice.

Identification of Pctaire-1 as a novel MRTF/SRF target gene in the brain

We show that the MRTF/SRF pathway regulates Pctaire-1 kinase in the brain. Pctaire-1 is an atypical Cdc-like kinase in that it does not require cyclins for activation and is not involved in cell cycle regulation (Besset et al., 1998; Graeser et al., 2002; Okuda et al.,

1992). It is expressed at low levels in most cell types, but is particularly abundant in postmitotic neurons (Besset et al., 1999). Two lines of evidence link Pctaire-1 function in neurons to regulation of neurite outgrowth; the first showing regulation of neurite outgrowth by Pctaire-1 in Neuro-2A cells (Graeser et al., 2002), and the second reporting the interaction between Pctaire-1 and the Cdk5 pathway (Cheng et al., 2002), which is implicated in regulation of diverse neuronal processes including regulation of actin dynamics, neuronal migration, and neurite outgrowth. We show here that MRTF/SRF activates Pctaire-1 gene expression through a CArG box upstream of the first exon of the gene, and that knockdown of Pctaire-1 blocks the growth of neurites in primary neuron cultures, mimicking the effect of MRTF deletion.

Cdk5 kinase interacts with its brain-specific coactivator p35 to drive neuronal differentiation, actin remodeling, neuronal migration, and neurite outgrowth (Dhavan and

44

Tsai, 2001). Cdk5 and p35 knockout mice have phenotypes that resemble some of the phenotypes observed in the MRTF bdKO mice as well as SRF knockout mice. Cdk5 phosphorylates and inhibits Pak1, linking the activity of Cdk5 to regulation of the actin cytoskeleton (Dhavan and Tsai, 2001; Smith, 2003). Pak1 can directly phosphorylate actin, resulting in the dissolution of stress fibers. Pak1 also increases the phosphorylation activity of LimK towards Cofilin, blocking its depolymerizing activity and thereby inhibiting actin turnover (Arber et al., 1998; Yang et al., 1998). Interestingly, we show that genetic deletion of MRTF-A and -B in the mouse brain causes dysregulation of the activity of Cdk5 accompanied by increased phosphorylation of LimK, possibly due to decreased phosphorylation and activation of Pak1. Disruption of this cascade of protein kinases results in hyperphosphorylation and inactivation of Cofilin in the brains of MRTF bdKO mice.

Overall, our findings and those of others demonstrate the essential role of the

MRTF/SRF signaling system in the coupling of cytoskeletal dynamics to nuclear gene transcription in the nervous system. Given the importance of MRTF/SRF in the control of cytoskeletal gene expression in other cell types, especially muscle cells, it will be interesting to determine whether the Pctaire-1/Cdk5 pathway identified in this study also modulates the actin cytoskeleton downstream of MRTF/SRF in other cell types.

METHODS

Mouse lines

The MRTF-B conditional null allele was generated using homologous recombination in embryonic stem cells. The pGKNEO-F2L2DTA vector, which contains a neomycin

45

resistance gene flanked by FRT and loxP sites and a diphtheria toxin gene cassette, was used for MRTF-B targeting. The 5’ arm, knockout arm, and 3’ arm of the targeting construct were generated by high-fidelity PCR amplification (Roche Expand High-

Fidelity Long Template) of 129SvEv genomic DNA. The targeting vector was linearized with PvuI and electroporated into 129SvEv-derived ES cells. Isolated ES cell clones were analyzed for homologous recombination. Incorporation of the 5’ and 3’ loxP sites was confirmed by Southern blotting using 5’ and 3’ probes following digestion with XbaI and SacI, respectively. Clones with the targeted MRTF-B allele were injected into 3.5- day C57BL/6 blastocysts, and the resulting chimeras were crossed to C57BL/6 females to achieve germline transmission of the targeted (MRTF-Bneo-loxP) allele. The MRTF-Bneo- loxP mice were crossed to Flpe transgenic mice then to GFAP-Cre transgenic mice (Zhuo et al., 2001) to obtain the MRTF-BF and MRTF-BKO alleles, respectively. MRTF-

BF/F;GFAP-Cre mice were then crossed to MRTF-A-/- mice (Li et al., 2006) to generate

MRTF-A-/-;MRTF-BF/F;GFAP-Cre (MRTF bdKO) mice.

Histology and Immunohistochemistry

Control and bdKO mice were anesthetized and transcardially perfused with PBS, followed by 4% paraformaldehyde prior to brain dissection. Brains were then postfixed in 4% paraformaldehyde for two days, embedded in paraffin, and sectioned. Sections were stained with hematoxylin and eosin or with Nissl stain using standard procedures

(Shelton et al., 2000). TUNEL assay and Golgi staining were performed according to manufacturers’ instructions (Roche and FD Neurotechnologies, respectively).

Immunohistochemistry using monoclonal anti-BrdU antibody (Roche), monoclonal anti-

46

MAP-2 antibody (Sigma), anti-Pctaire-1 antibody (Santa Cruz Biotechnology), anti-ph-

Cdk5 (Santa Cruz Biotechnology), anti-ph-LimK (Abcam), anti-ph-Cofilin (Sigma), and anti-Tubulin-J1 (Abcam) was performed using standard protocols.

BrdU labeling

Timed matings were set up between MRTF-A-/+;MRTF-BF/F female and MRTF-A-/+;

MRTF-BF/F;GFAP-Cre male mice. Pregnant females were IP injected with BrdU

(Roche) (100 μg/g body weight) at E15.5. The pups were genotyped and sacrificed at P7.

MRTF-A-/-;MRTF-BF/F; GFAP-Cre and MRTF-A-/+;MRTF-BF/F control brains were sectioned for hematoxylin and eosin staining and for BrdU labeling.

Neurite outgrowth assay

MRTF-A-/-;MRTF-BF/F and MRTF-A-/+;MRTF-BF/F mouse pups (P1-P3) were used for hippocampal and cortical cultures as described by (Ahlemeyer and Baumgart-Vogt,

2005)). The cultures were plated on poly-D-lysine/laminin coated coverslips (BD

Biosciences) and infected with either Ad5CMVCre-eGFP or Ad5CMVeGFP adenovirus constructs (University of Iowa Gene Transfer Vectore Core). After 2 weeks, the cells were fixed with 4% paraformaldehyde, permeabilized, and blocked with 3% bovine serum albumin in PBST buffer. The cells were then incubated with anti-MAP-2 primary antibody (1:500 in PBST) for 45min followed by 3 washes in PBST and incubated with

Texas-Red-conjugated secondary antibody. After 3 washes in PBST, coverslips were mounted on glass slides using VectaShield mounting medium with DAPI (Vector

Laboratories), and visualized with a confocal microscrope.

47

RT-PCR analysis

Total RNA was purified from tissues using TRIzol reagent according to the manufacturer’s instructions (Invitrogen). For RT–PCR, total RNA was used as a template for RT using random hexamer primers. Quantitative real-time PCR was performed using TaqMan probes purchased from ABI.

Western blots

Brain tissues were homogenized in lysis buffer (50 mM Tris at pH 7.4, 150 mM NaCl,

1% Triton X-100, 1 mM EDTA) supplemented with protease inhibitors (Complete Mini,

EDTA-free, Roche) and phosphatase inhibitors (Sigma). The lysates were then centrifuged at 14,000g for 5 min, and supernatants were recovered. Equal amounts of protein extracts were resolved by SDS-PAGE on a 12% acrylamide gel and analyzed by

Western blot using primary antibody for ph-Cofilin (Sigma), total Cofilin (Sigma) and

GAPDH (Millipore), followed by the corresponding IgG HRP-conjugated secondary antibody (Bio-Rad) and detected by enhanced chemiluminescence (Western Blot

Luminol Reagent, Santa Cruz Biotechnology).

Gel mobility shift assays

GST-SRF and control GST proteins were purified from bacterial E-coli cultures using standard protocols. Wild-type and mutant DNA probes were labeled with 32P-dCTP using a Klenow fill-in method and purified using a G25 DNA purification column

(Roche). The 20 μl binding reaction contained 1 μg of poly (dI-dC) (Sigma), 100,000

48

cpm of probe, and increased amounts of purified GST-SRF. After incubation at room temperature for 15 min, the reactions were separated on a 5% PAGE gel containing 0.5 ×

TBE.

Luciferase assays

The Pctaire-1-luciferase construct contained a 500bp DNA fragment located upstream of the starting codon of the Pctaire-1 gene. Cos cells were plated in 24-well plates (5x104 cells per well) and transfected with 150 ng of the Pctaire-1-luciferase construct together with 50ng of the expression plasmids encoding Myocardin and 10, 50, and 100ng of the

SRF expression plasmid using 1.4 μl of FuGENE 6 reagent (Roche Molecular

Biochemicals). Transfection efficiency was normalized by cotransfection of 10 ng of pCMV-LacZ. 48 h posttransfection, the cells were harvested in 150 μl of passive lysate buffer (Promega), and 20 μl of cell lysate was used for luciferase or β-galactosidase assays.

Kinase assay

Cdk5 was immunoprecipitated from control or MRTF bdKO cortices using anti-Cdk5 antibody (Santa Cruz Biotechnology), conjugated to glutathione-agarose beads, and washed with PBS. Immunoprecipitated Cdk5 beads were then resuspended in kinase reaction buffer (30 μl) containing 12.5 μM ATP and 5 μCi of [γ-32P]-ATP, and recombinant histone H1 protein (100ug). Reactions were allowed to proceed for 30 min at room temperature and phosphoproteins were then bound to whatman filter papers and analyzed by Beckman counter.

49

Statistical analysis

Results are expressed as means ± SEM. Unpaired 2-tailed Student t-test with Welch correction was performed to determine statistical significance between groups. P values of <0.05 were considered significant.

50

Chapter III

A threshold of MRTFs is Required for

Cardiac Development and Function

51

ABSTRACT

MRTFs are powerful transcriptional activators that modulate SRF activity in muscle cells.

SRF plays an essential role in development and maintenance of cardiac function. MRTF-

A and -B possess highly conserved domain structures, are coexpressed with SRF in the embryonic and adult heart, and are able to redundantly coactivate SRF in vitro. Here, we generated mice lacking both MRTF-A and -B in the heart to elucidate the in vivo functions of MRTFs in cardiac development. We show that cardiac deletion of MRTFs causes a range of cardiac defects, whose severity correlates with the number of deleted

MRTF alleles, and that range from reduced cardiac contractility to neonatal lethality with major defects in sarcomere arrangement. Importantly, we identify the gene encoding

Pctaire-1, a Cdc2-like serine/threonine-specific protein kinase, never before studied in the heart, as a regulator of sarcomere arrangement and an effector of the functions of

MRTF-A and -B in cardiogenesis.

51

INTRODUCTION

Formation of the heart and maintenance of cardiac function are complex processes that are subject to tight regulation. Minor alterations in these processes have profound consequences and can result in congenital and acquired cardiovascular diseases.

Deciphering the gene regulatory networks governing the development and maintenance of cardiac function is thus crucial for further understanding of cardiovascular disease.

Emerging evidence has revealed the role of SRF in cardiogenesis and cardiac remodeling using several strains of mice with cardiac specific SRF gene knockouts.

Deletion of SRF in the embryonic heart, using either beta-MHC-Cre or alpha-MHC-Cre transgenic mice, impairs cardiac differentiation and causes cardiac insufficiency, respectively. These SRF deletions disrupt the expression of multiple regulators of cardiac development, such as Nkx2.5, GATA4, and ANF as well as cardiac structural components such as cardiac, skeletal, and smooth muscle alpha-actin proteins (Niu et al.,

2005; Parlakian et al., 2004). Moreover, deletion of SRF in the adult heart, using heart- specific tamoxifen-inducible Cre mice, causes impairment of left ventricular function, dilated cardiomyopathy, and heart failure, demonstrating the requirement for SRF for adult cardiac function and integrity (Parlakian et al., 2005).

MRTFs are major regulators of SRF activity in muscle cells in vitro. Expression of a dominant negative MRTF mutant in C2C12 myoblasts blocks the expression of SRF target genes and prevents myogenic differentiation into myotubes (Selvaraj and Prywes,

2003). Moreover, our preliminary studies using MRTF null MEFs revealed a redundant role for MRTF-A and -B in the regulation of SRF mediated signaling in vitro, supporting earlier studies where deletion of both MRTF-A and -B was necessary to block Rho

52

mediated activation of SRF in HeLa cells (Chapter II of this dissertation) (Cen et al.,

2003).

In vivo, MRTF-A and -B are coexpressed with SRF in the embryonic and adult heart. Studies of various mutants of Drosophila MRTF support an essential and redundant function for DMRTF as a regulator and essential partner of Drosophila SRF

(Han et al., 2004). However, whether this partnership is conserved to mammals is still questionable, especially since no cardiac phenotype was observed with any of the single

MRTF knockout mice (Li et al., 2005a; Li et al., 2006; Li et al., 2003; Oh et al., 2005).

The objective of the following experiments is thus to decipher a putative role for MRTFs in cardiac development and remodeling and to test their requirement for SRF activity in muscle cells in vivo. We show that cardiac deletion of MRTF-B in an MRTF-A heterozygous background causes reduced cardiac contractility and partial lethality.

Moreover, ablation of MRTF-B in an MRTF-A null background causes neonatal lethality with defects in sarcomere arrangement and abnormal expression of numerous SRF target genes encoding sarcomeric and cytoskeletal proteins. Moreover, we identify the gene encoding Pctaire-1 protein kinase as a novel SRF target gene and regulator of sarcomere assembly in the heart. Using Pctaire-1 siRNA knockdown and alpha-MHC-Pctaire-1 transgenic mice, we demonstrate a previously undescribed role for Pctaire-1 as a mediator of MRTF/SRF signaling and regulator of sarcomere arrangement.

RESULTS

Generation of mice lacking MRTF-A and MRTF-B in the heart

53

To explore the potential functions of MRTFs in cardiac development, we generated mice lacking both MRTF-A and MRTF-B in the heart. Using MRTF-AKO and MRTF-BF/F mice, we deleted MRTFB in the hearts of MRTF-AKO mice using alpha-MHC-Cre transgenic mice. We refer to the MRTF-AKO; MRTF-BF/F; alpha-MHC-Cre mice as

MRTF cardiac double knockout mice (MRTF cdKO). MRTF cdKO mice were born at

Mendelian ratios, but 75% of these mice died at postnatal day 1 (P1). The other 25% of the mice die gradually between weeks 4 and 12 (Figure 3.1). Quantitative real-time PCR, using primers located within the deleted exons of either MRTF-A and MRTF-B, confirmed MRTF takeout in the MRTF cdKO hearts compared to control hearts (Figure

3.1). Interestingly, 25% of the MRTF-Ahet;MRTF-BcKO, which have one functional

MRTF-A allele in the heart, died between weeks 2 and 4 (Figure 3.1). We also observed partial lethality of MRTF-AKO; MRTF-BF/+; alpha-MHC-Cre mice, suggesting that

MRTF-A and -B have redundant functions in the heart and that and that a threshold of

MRTF gene expression is required to support normal cardiac development and function.

Phenotype of mice lacking MRTF-B in an MRTF-A null background.

Analyses of mice lacking MRTF-B in an MRTF-A null background by hematoxylin and eosin (H&E) staining revealed a range of cardiac phenotypes. 75% of the hearts of

MRTF-Ahet; MRTF-BcKO mice appeared normal and indistinguishable from the control hearts. However, in correlation with the percent of lethality of these mice, the other 25% of the hearts showed cardiac dilation, endocardial fibrosis, and disarrangement of cardiomyocytes. As with MRTF-Ahet;MRTF-BcKO mice, the MRTF cdKO mice showed

54

Figure 3.1. Generation of MRTF cdKO mice. (A) Quantitative RT-PCR analysis of MRTFs in MRTF cdKO hearts. (B) Survival curves for mice with cardiac deletion of MRTF-A and MRTF-B. One copy of MRTF-A results in 25% lethality by 4 weeks of age, whereas 80% of MRTF cdKO mice exhibit perinatal lethality.

55

massive cardiac dilation, endocardial fibrosis, and disarrangement of cardiomyocytes

(Figure 3.2).

To assess cardiac functionality of MRTF-Ahet; MRTF-BcKO hearts, we performed echocardiography, measured the left ventricular internal diameter at diastole and systole, and calculated the percent fractional shortening. MRTF-Ahet;MRTF-BcKO hearts showed a significant decrease in their fractional shortening reflecting their compromised contractility and function (Figure 3.2).

Defective sarcomere arrangement in MRTF cdKO hearts.

We examined the hearts of the MRTF cdKO mice at P1, the point of maximal lethality.

MRTF cdKO hearts appeared normal and indistinguishable from control hearts whether by whole mount or by H&E (Figure 3.3). Interestingly, however, the MRTF cdKO hearts showed dramatic disruption of sarcomeres by electron microscopy with completely disorganized Z-bands and barely recognized M-bands compared to control hearts (Figure

3.3). The MRTF cdKO cardiomyocytes also contained misshapen mitochondria, but we are currently uncertain whether this defect is a primary or secondary effect of MRTF deletion.

Molecular dissection of the MRTF cdKO phenotype

To gain further insight into the cause of lethality of MRTF cdKO mice at birth, we performed microarray analysis on the hearts of these mice. Many of the genes that were significantly downregulated and that we were able to confirm by quantitative real-time

PCR analyses are target genes of the SRF/MRTF pathway and encode sarcomeric

56

Figure 3.2. A range of cardiac defects upon MRTF deletion in the heart. (A) H&E stained heart sections from MRTF-Ahet;MRTF-BcKO mice and MRTFcdKO mice of the indicated genotypes at 3 weeks are shown at low and high magnification in the upper and lower panels, respectively. About 25% of MRTF-Ahet;MRTF-BcKO and the surviving MRTF cdKO mice show cardiac dilation and endocardial fibrosis with incomplete penetrance. (B) Echocardiograms from mice of the indicated genotypes are shown. (C) Quantitative analyses of the echocardiograms from mice of the indicated genotypes are shown. Left ventricular internal diameter at diastole (LVIDd), at systole (LVIDs) and fractional shortening (FS) data are shown.

57

Figure 3.3. Defects in sarcomere arrangement in MRTF cdKO hearts. (A) Whole mount (left panel) and hematoxylin-eosin stained heart sections (right panel) from control and MRTF cdKO mouse neonates are shown. (B) Electron micrographs of control hearts and MRTF cdKO hearts at birth reveal disorganization of the sarcomeres.

58

proteins such as the cardiac and smooth muscle alpha-actins (Acta-1 and -2), myosin light chain-1(Myl1) and the myosin light chain kinase-4 (Mylk-4), tropomyosins-1 and -2

(Tpm-1and -2), and calponin (Cnn1) (Figure 3.4). Even though many of these sarcomeric proteins are known to have long half lives, we were able to confirm downregulation of the phosphorylated form of MLC, smooth muscle alpha-actin, and calponin (Figure 3.4).

Pctaire-1 in cardiac development

In addition to sarcomere genes, the gene encoding the protein kinase Pctaire-1, which we showed to be a target of the MRTF/SRF pathway in the brain (chapter II of this dissertation), was also among the most downregulated genes retrieved by microarray.

To gain more insights into the putative functions of Pctaire-1 in the heart, we started by examining the cellular localization of Pctaire-1 in rat neonatal cardiomyocytes.

We were able to detect Pctaire-1 in cardiomyocytes by immunohistochemistry (Figure

3.5). Pctaire-1 was also expressed at a lower level in cardiac fibroblasts (data not shown).

Upon overexpression, Pctaire-1 was detected in multiple cellular structures, including vesicular, sarcomeric, focal adhesion, and golgi localization (Figure 3.5).

Gene expression analysis by quantitative real-time PCR confirmed diminished expression of Pctaire-1 in the hearts of MRTF cdKO mice (Figure 3.5). Furthermore, we confirmed diminished expression of Pctaire-1 proteins in the hearts of MRTF cdKO hearts compared to control mice by western blots, suggesting that Pctaire-1 is a target of

MRTF/SRF signaling in the heart (Figure 3.5). Interestingly, we show by western blot diminished phosphorylation of Cofilin, a downstream regulator of actin polymerization,

59

Figure 3.4. Dysregulation of sarcomere components in MRTF cdKO hearts. (A) Analysis of sarcomere gene expression in MRTF cdKO hearts at birth. Microarray data were confirmed by quantitative RT-PCR showing downregulation of the sarcomere genes skeletal and smooth muscle alpha-actins (Acta1 and Acta2), myosin light chain (Myl1), myosin light chain kinase (mylk4), calponin (Cnn1), and tropomyosins 1 and 2 (Tpm1 and Tpm2). (B) Western blot analysis for control (lane1), MRTF-Ahet;MRTF-BcKO (lane 2), and MRTF cdKO hearts (lane3). Phosphorylated myosin light chain (ph-MLC), smooth muscle a-actin (SM-a-actin) and calponin (Cnn1) are downregulated in MRTF cdKO hearts. GAPDH was used as loading control.

60

Figure 3.5. Pctaire-1 as a mediator of MRTF/SRF signaling in the heart. (A) Expression and localization of Pctaire-1 in rat neonatal cardiomyocytes. (B) Quantitative RT-PCR showed decreased expression of Pctaire-1 transcripts in MRTF cdKO hearts. (C) Western blot showed diminished levels of Pctaire-1 proteins in MRTF cdKO hearts. (D) Increased Cofilin phosphorylation in MRTF cdKO hearts shown by western blot. (E,F) Disruption of sarcomeres upon knockdown of Pctaire-1 in cardiomyocytes. (E) Quantitative RT-PCR showed significant knockdown of Pctaire-1 by siRNAs 1 and 2. (F) Sarcomeric alpha-actinin staining showed disruption of the sarcomeres in cardiomyocytes transfected with siRNAs 1 and 2, but not in cardiomyocytes transfected with non-targeting control siRNA.

61

in the hearts of MRTF cdKO mice compared to control hearts (Figure 3.5), suggesting that the molecular mechanisms we described in the brain (chapter II of this dissertation) are conserved in the heart.

Next, we knocked down Pctaire-1 in neonatal rat cardiomyocytes using three different siRNAs. We confirmed knockdown of Pctaire-1 by two different siRNAs by quantitative RT-PCR (Figure 3.5). Interestingly, staining of Pctaire-1 knockdown cardiomyocytes with sarcomeric alpha-actinin showed abnormal cellular morphology with disruption of sarcomere arrangement (Figure 3.5), suggesting that Pctaire-1 activity is required for normal sarcomere assembly in cardiomyocytes.

To examine the in vivo functions of Pctaire-1 in the mouse heart, we generated multiple transgenic mouse lines that overexpress HA-tagged Pctaire-1 under the control of the alpha-MHC promoter. We are currently analyzing two different lines that show different levels of Pctaire-1 overexpression as shown by western blots against HA and

Pctaire-1 (Figure 3.6). Western blot against the phosphorylated form of cofilin suggests increased phosphorylation of Cofilin upon overexpression of Pctaire-1 (Figure 3.6).

Interestingly, H&E stained sections of alpha-MHC-Pctaire-1 hearts at 6 weeks of age

(from Line 1) show thinning of the myocardium and disruption of cardiomyocyte morphology, supporting a role for Pctaire-1 in the regulation of cardiac development

(Figure 3.6). We are currently analyzing the molecular and morphological phenotype of

Pctaire-1 transgenic mice in Line 3 to further decipher the functions of Pctaire-1 in the heart.

62

Figure 3.6. Regulation of cardiac development in vivo. (A) Generation of alpha- MHC-HA-Pctaire-1 transgenic mice. Western blots show two lines of transgenic mice expressing different levels of the transgene in anti-HA and anti-Pctaire-1 blots and increased Cofilin phosphorylation by anti-ph-Cofilin blotting. GAPDH was used as loading control. (B) Cardiac dilation upon overexpression of Pctaire-1 in the heart. Shown are H&E stained sections of 6 weeks old hearts from control and transgenic mice (Line 1).

63

DISCUSSION

The results of this study demonstrate the requirement for a threshold of MRTF gene level during cardiac development and function. Deletion of three MRTF alleles compromises cardiac function and causes dilated cardiomyopathy and eventually heart failure.

Ablation of all four MRTF alleles causes perinatal lethality with disruption of sarcomeric gene expression. These results also identify a previously unidentified role for Pctaire-1 as a mediator of MRTF/SRF signaling in the heart and a regulator of cardiac development.

Redundant functions of MRTFs in the heart

This study reveals that MRTF-A and -B function redundantly during cardiac development and function. Unlike the brain, the heart seems to be more sensitive to

MRTF gene dosage. Deletion of three MRTF alleles has no overt effect on brain development, but reduces cardiac functionality and causes partial lethality before adulthood when deleted in the heart. This suggests that the heart might be more sensitive to MRTF mediated signaling than the brain. Alternatively, this difference could be attributed to different onsets of expression of MRTF-A and -B between the brain and the heart. Therefore a more detailed analysis of the onset and level of expression of each

MRTF in various tissues would complement the data obtained from MRTF dKO mice to further discern between MRTF redundant and specific functions in various tissue contexts.

64

Figure 3.7. Model for MRTF function during cardiac development. MRTFs regulate the expression of numerous sarcomeric genes including actins, tropomyosins, and calponin. MRTFs also regulate the expression of Pctaire-1, which itself is a regulator of sarcomere assembly and cardiac development.

65

MRTFs in SRF signaling

The phenotype observed with the MRTF cdKO mice phenocopies some aspects of the defects observed with the cardiac deletion of SRF. Mice bearing both genotypes succumb to death with disrupted sarcomere organization and dysregulated expression of several SRF target genes. However, unlike the alpha-MHC-Cre SRF knockout mice, which die embryonically by E12.5, MRTF cdKO mice die around P1 (Niu et al., 2005).

Even though we cannot exclude the possibility that other SRF coactivators contribute to the earlier lethality with the cardiac SRF deletion, it is also likely that Myocardin compensates to some extent for the loss of MRTF activity between E12.5 and P1. In fact,

Myocardin is coexpressed with MRTF-A and -B in the embryonic heart. And even though the hearts of Myocardin knockout mice seem normal until E10.5, when they die from defective smooth muscle differentiation, a role for Myocardin in cardiac development is still debatable. MASTR, on the other hand, is expressed in the heart but is not likely to compensate for MRTF function as it lacks the SRF binding domain and is thought to function exclusively as an activator of MEF-2.

Functions of Pctaire-1 in cardiac development

As stated earlier, Pctaire-1 was originally identified as a Cdc2-like serine/threonine- specific protein kinase, which does not require cyclins for activation and is not involved in cell cycle regulation (Besset et al., 1998; Graeser et al., 2002; Okuda et al., 1992). It is expressed at low levels in most cell types, but particularly abundant in terminally differentiated cells (Besset et al., 1999). Pctaire-1 has been linked to Cdk5 signaling in neurons and suggested to be involved in neurite outgrowth in Neuro-2A cells (Cheng et

66

al., 2002; Graeser et al., 2002); however Pctaire-1 has no known cardiac function. We show here that the transcript and protein levels of Pctaire-1 are diminished upon cardiac deletion of MRTFs, suggesting that expression of Pctaire-1 is partially dependent on

MRTF/SRF signaling in the heart. Consistent with its essential role as an effector of the functions of MRTF-A and -B in cardiogenesis, knockdown of Pctaire-1 expression in cardiomyocytes results in sarcomere disassembly, similar to the defects seen in MRTF-

A/-B double mutant mice. Moreover, overexpression of Pctaire-1 in the mouse heart disrupts cardiomyocyte morphology and results in thinning of the myocardium at 6 weeks of age, further supporting and essential role for Pctaire-1 in the regulation of cardiac cytoskeleton and development in vivo. We are currently analyzing hearts from mice younger and older than 6 weeks to examine the onset and progression of the phenotype.

As mentioned earlier, Pctaire-1 was reported to interact with Cdk5 in neurons

(Chen et al., 2007; Cheng et al., 2002; Graeser et al., 2002) . In addition to its involvement in various neuronal processes, Cdk5 is also involved in neuromuscular and myogenic signalling (Bajaj, 2000; Dhavan and Tsai, 2001). The levels and activity of

Cdk5 have been shown to increase during early myogenesis of mouse C2 myoblasts and decrease during myotube fusion. Expression of wild type or dominant negative Cdk5 results in enhanced and diminished differentiation of C2 myoblasts into myotubes, respectively (Lazaro et al., 1997). Moreover, expression of dominant negative Cdk5 in

Xenopus embryos disrupts somatic muscle patterning and suppresses the expression of the myogenic MYOD and MRF4 transcription factors (Philpott et al., 1997). Therefore, it would be interesting to assess the expression and activity of Cdk5 in MRTF cdKO hearts and in Pctaire-1 knockdown cardiomycytes.

67

Based on the above, we conclude that MRTF-A and -B act redundantly as major regulators of cardiac development and function. Cardiac deletion of MRTFs results in downregulation of numerous known SRF target genes encoding sarcomere components.

In addition, deletion of MRTFs results in downregulation of Pctaire-1, a mediator of

MRTF/SRF signaling and regulator of sarcomere assembly.

METHODS

Mouse lines

The MRTF-B conditional null allele was generated using homologous recombination in embryonic stem cells as described in chapter II of this dissertation. MRTF-A knockout mice were used (Li et al., 2006). MRTF-BF/F mice (described in chapter II of this dissertation) were crossed to MRTF-AKO mice (Li et al., 2006) to generate MRTF-AKO;

MRTF-BF/F; alpha-MHC-Cre (MRTF cdKO) mice.

Histology, immunohistochemistry, and electron microscopy

Control and cdKO mice were sacrificed at the indicated time points and fixed in 4% paraformaldehyde for two days, embedded in paraffin, and sectioned. Sections were stained with hematoxylin and eosin stain using standard procedures (Shelton et al., 2000).

For transmission electron microscopy, left ventricular heart tissue was minced and fixed in 2% paraformaldehyde, 2.5% glutaraldehyde, and 0.1 M cacodylate buffer, prepared according to standard protocol; and electron microscopy was performed at the University of Texas Southwestern Molecular and Cellular Imaging Facility using a Tecnai G2 Spirit

120 KV TEM.

68

Cell culture and immunocytochemistry

Rat neonatyal cardiomyocytes were isolated using standard protocols. The cultures were plated on poly-D-lysine/laminin coated coverslips (BD Biosciences) and transfected with

Pctaire-1 siRNA and control siRNA (Dharmacon) using lipofectamineTM 2000 reagent

(Invitrogen) according to the manufacturer’s protocol. After 2 days, the cells were fixed with 4% paraformaldehyde, permeabilized, and blocked with 3% bovine serum albumin in PBST buffer. The cells were then incubated with anti-sarcomeric-alpha-actinin antibody (Sigma) for 45min followed by 3 washes in PBST and incubated with Texas-

Red-conjugated secondary antibody. After 3 washes in PBST, coverslips were mounted on glass slides using VectaShield mounting medium with DAPI (Vector Laboratories), and visualized with a confocal microscrope.

Quantitative RT-PCR analysis

Total RNA was purified from tissues using TRIzol reagent according to the manufacturer’s instructions (Invitrogen). For RT–PCR, total RNA was used as a template for RT using random hexamer primers. Quantitative real-time PCR was performed using TaqMan probes purchased from ABI.

Western blots

Cortical brain tissues were homogenized in lysis buffer (50 mM Tris at pH 7.4, 150 mM

NaCl, 1% Triton X-100, 1 mM EDTA) supplemented with protease inhibitors (Complete

Mini, EDTA-free, Roche) and phosphatase inhibitors (Sigma). The lysates were then

69

centrifuged at 14,000g for 5 min, and supernatants were recovered. Equal amounts of protein extracts were resolved by SDS-PAGE on a 12% acrylamide gel and analyzed by

Western blot using primary antibody for ph-MLC (Santa Cruz Biotechnology), SM- alpha-actin (Sigma), Cnn1 (Sigma), Pctaire-1 (Santa Cruz Biotechnology), and GAPDH

(Millipore), followed by the corresponding IgG HRP-conjugated secondary antibody

(Bio-Rad) and detected by enhanced chemiluminescence (Western Blot Luminol Reagent,

Santa Cruz Biotechnology).

Transthoracic echocardiography

Cardiac function and heart dimensions were evaluated by 2-dimensional echocardiography on conscious mice using a Vingmed System (GE Vingmed Ultrasound) and a 11.5-MHz linear array transducer. M-mode tracings were used to measure anterior and posterior wall thicknesses at end diastole and end systole. LV internal diameter

(LVID) was measured as the largest anteroposterior diameter in either diastole (LVIDd) or systole (LVIDs). The data were analyzed by a single observer blinded to mouse genotypes. LV fractional shortening (FS) was calculated according to the following formula: FS (%) = [(LVIDd – LVIDs)/LVIDd] × 100.

Statistical analysis

Results are expressed as means ± SEM. Unpaired 2-tailed Student t-test with Welch correction was performed to determine statistical significance between groups. P values of <0.05 were considered significant.

70

Chapter IV

MASTR Cooperates with MRTF-A to

Control Skeletal Muscle Development

71

ABSTRACT

MASTR is a Myocardin related protein highly enriched in skeletal muscle and brain.

MASTR and Myocardin share homology with a MEF2 binding motif and a SAP domain, and both funcion as strong transcriptional activators. Studies performed in cell culture models and Xenopus embryos suggest that MASTR functions as a promyogenic protein.

To test this hypothesis, we generated a MASTR null allele in mice. MASTR knockout mice were born at Mendelian ratios and do not show any significant phenotype.

However, deletion of MASTR and MRTF-A causes perinatal lethality, possibly due to defective skeletal muscle development. These findings suggest that MASTR and MRTF-

A play redundant roles in muscle development, possibly mediated by associations with cofactors through their homologous SAP domains.

72

INTRODUCTION

MASTR is an extremely powerful transcriptional activator highly enriched in brain and skeletal muscle. MASTR, which was identified by a BLAST search for Myocardin related ESTs, is a 421 amino acid protein with three known domains; an N-terminal motif homologous to the MEF2 binding domain of the cardiac Myocardin isoform, a SAP domain, and a transactivation domain (Figure 4.1). MASTR, which localizes exclusively to the nucleus, binds MEF2 and potentiates its transcriptional activity, but is not able to activate SRF dependent gene transcription. In vitro, MASTR is able to enhance the myogenic activity of MyoD in 10T1/2 fibroblasts, which are able to differentiate into myotubes under promyogenic culture conditions (Creemers et al., 2006). MASTR is not a direct activator of MyoD, and its effect of myogenesis of 10T1/2 cells is thought to be indirect, possibly mediated by MEF2. In Xenopus embryos, knockdown and overexpression of MASTR were shown to modulate the expression of skeletal muscle marker genes, further suggesting its role in muscle development (Meadows et al., 2008).

MEF2 proteins have been shown to control growth and differentiation in different cell types, particularly in muscle cells (McKinsey et al., 2002; Potthoff and Olson, 2007).

MEF2C for instance is required for cardiovascular development in mouse embryos. In the adult heart, MEF2 proteins are also required to modulate cardiac function under normal and pathological conditions by regulating the expression of genes involved in cardiac contractility and energy metabolism (Kim et al., 2008; Lin et al., 1998; Lin et al.,

1997; Naya et al., 2002). In skeletal muscle, MEF2C was also shown to be indispensable for postnatal muscle maturation, fiber type specification, and maintenance of sarcomeric integrity. Skeletal muscle specific deletion of MEF2C causes disorganization of

73

Figure 4.1. Expression analysis of MASTR. (A) Domain structure of MASTR. The homology between the MEF2 binding domain and SAP domain of MASTR and Myocardin is shown. (B) Quantitative RT-PCR analysis of the expression of MASTR on days 0 (d0) to 5 (d5) of C2C12 myoblast differentiation. (C) Quantitative RT-PCR analysis of the expression of MASTR in various adult brain structures and muscle tissues.

74

myofibers and perinatal lethality with disrupted sarcomere organization (Potthoff et al.,

2007a). Moreover, expression of a hyperactive MEF2 mutant in skeletal muscle enhances muscle endurance by promoting the formation of slow fibers (Potthoff et al.,

2007b). These studies suggest that MASTR could function in vivo to control MEF2 activity in muscle cells.

To explore the in vivo contributions of MASTR to myogenesis, we generated

MASTR knockout mice, which do not show any obvious phenotype. However, ablation of MASTR in an MRTF-A null background is lethal between P1 and P14, possibly due to skeletal muscle defects. Analysis of the phenotype of MASTR/MRTF-A double knockout (dKO) mice is underway.

RESULTS

Expression analysis of MASTR

As stated earlier, MASTR was shown to be highly enriched in brain and muscle. We performed quantitative RT-PCR to further examine the expression of MASTR in various brain regions and muscle types. We show that MASTR expression is most prominent in the cerebellum and skeletal muscle. MASTR transcripts are also enriched in the heart and hippocampus (Figure 4.1). Consistent with a possible role for MASTR in myogenesis, we show by quantitative RT-PCR that expression of MASTR increases during differentiation of C2C12 myoblasts into myotubes and peaks on day 2 of differentiation (Figure 4.1).

Generation of MASTR knockout mice

75

To explore the potential in vivo functions of MASTR, we generated a global MASTR deletion in mice using a conditional MASTR allele, which was generated by introducing loxP sites within introns 1 and 7 of the mouse MASTR locus by homologous recombination in embryonic stem cells (Figure 4.2) (Collaboration with Esther

Creemers). Deletion of exons 2 through 7, which encode the MEF2 binding motif, SAP domain, and part of the transactivation domain, eliminates the transcriptional activity of

MASTR in vitro (Creemers et al., 2006), thereby generating a MASTR null allele. The

MASTRFloxed mice were bred to CAG-Cre transgenic mice, which express Cre recombinase globally in the mouse embryo. Correct targeting and germline transmission were confirmed by Southern blot and PCR analyses of genomic DNA. Deletion of

MASTR was confirmed by RT-PCR from skeletal muscle RNA of control and MASTR knockout (KO) skeletal muscle tissues (Figure 4.2). MASTR KO mice were phenotypically normal and displayed no obvious phenotype.

Generation of MASTR/MRTF-A double knockout mice

To examine possible redundancy between MASTR and other MRTF proteins, we made use of MRTF-A knockout mice to generate MRTF-A/MASTR double KO (dKO) mice.

MRTF-AKO; MASTRhet and MRTF-Ahet; MASTRKO mice were phenotypically normal, suggesting that one copy of either MRTF-A or MASTR is sufficient to support normal development. However, deletion of both MRTF-A and MASTR results in lethality between P1 and P14 (Figure 4.3). Histological analysis of MRTF-A/MASTR dKO mice revealed multiple abnormalities reminiscent of a myopathic phenotype. H&E staining of dKO skeletal muscle at P14 showed heterogeneity and reduction of myofiber size with

76

Figure 4.2. Generation of MASTR knockout mice. (A) Strategy to generate a MASTR knockout mice. Protein, corresponding exonic structure, and targeted alleles are shown. (B) Southern blot analysis confirming targeting of ES cells. The corresponding wild type and targeted bands are indicated for the 5’ and 3’ probes. (C) Genotyping of wild-type, heterozygous, and knockout mice by genomic PCR. (D) Quantitative RT-PCR confirms deletion of MASTR in MASTR KO mice.

77

Figure 4.3. Defective skeletal muscle development in MRTF-A/MASTR dKO mice. (A,B) Lethality of MRTF-A/MASTR dKO mice between P1 and P14. (C) Myopathy of skeletal muscle in MRTF-A/MASTR dKO mice. H&E stained sections show heterogeneity of fiber size and nuclear fragmentation in dKO mice.

78

centralized and fragmented nuclei, suggesting an essential role for MRTF-A and MASTR in skeletal muscle development (Figure 4.3).

DISCUSSION

The results of this study are the first to reveal an in vivo function for MASTR in mouse development. Deletion of MASTR alone did not cause any overt phenotype; however, deletion of MASTR and MRTF-A in mice results in postnatal lethality between P1 and

P14, possibly due to skeletal muscle defects. Further studies are ongoing to dissect the phenotype at the morphological and molecular levels.

MASTR was originally cloned as a strong activator of MEF2 and hypothesized to promote myogenesis by MEF2 activation (Creemers et al., 2006). We had hypothesized that MASTR deletion disrupts the activity of MEF2, especially in skeletal muscle where

Myocardin is not expressed. However, MASTR knockout mice exhibit normal skeletal development and fiber type specification (data not shown). The skeletal muscle defects observed upon additional deletion of MRTF-A, which itself is not a MEF2 activator, suggest that MASTR functions are not exclusively MEF2 dependent or that MASTR and

MRTF-A function cooperatively or in parallel in a pathway that does not involve MEF2.

The only region of amino acid sequence homology between MASTR and MRTF-A is within the SAP domain, which has been implicated in the control of chromatin structure and protein-protein interactions. Thus, it is tempting to speculate that MASTR and

MRTF-A interact with a common partner protein through their SAP domains, thereby playing redundant roles in muscle maturation. Future studies will explore this hypothesis.

79

On the other hand, the skeletal muscle defects observed in MRTF-A/MASTR dKO mice resemble to some extent the phenotype observed upon skeletal muscle deletion of SRF. Ablation of SRF using MCK-Cre transgenic mice causes perinatal lethality with severe skeletal muscle hypoplasia (Li et al., 2005b). Therefore, it will be interesting to examine whether MASTR contributes to SRF activation in skeletal muscle. It will also be interesting to determine whether MASTR modulates the transcriptional activity of other transcription factors during myogenesis.

METHODS

Mouse lines

The MASTR conditional null allele was generated using homologous recombination in embryonic stem cells. The pGKNEO-F2L2DTA vector, which contains a neomycin resistance gene flanked by FRT and loxP sites and a diphtheria toxin gene cassette, was used for MASTR targeting. The 5’ arm, knockout arm, and 3’ arm of the targeting construct were generated by high-fidelity PCR amplification (Roche Expand High-

Fidelity Long Template) of 129SvEv genomic DNA. Isolated ES cell clones were analyzed for homologous recombination. Incorporation of the 5’ and 3’ loxP sites was confirmed by Southern. Clones with the targeted MASTR allele were injected into 3.5- day C57BL/6 blastocysts, and the resulting chimeras were crossed to C57BL/6 females to achieve germline transmission of the targeted (MRTF-Bneo-loxP) allele. The MRTF-Bneo- loxP mice were crossed to CAG-Cre transgenic mice to obtain the MASTRKO allele.

MASTRKO mice were then crossed to MRTF-AKO mice (Li et al., 2006) to generate

MRTF-A/MASTR double knockout (dKO) mice.

80

Histology, immunohistochemistry, and electron microscopy

Control and dKO mice were sacrificed and skeletal muscle fixed in 4% paraformaldehyde for two days, embedded in paraffin, and sectioned. Sections were stained with hematoxylin and eosin stain using standard procedures (Shelton et al., 2000).

Quantitative RT-PCR analysis

Total RNA was purified from cells or tissues using TRIzol reagent according to the manufacturer’s instructions (Invitrogen). For RT–PCR, total RNA was used as a template for RT using random hexamer primers. Quantitative real-time PCR was performed using TaqMan probes purchased from ABI.

Statistical analysis

Results are expressed as means ± SEM. Unpaired 2-tailed Student t-test with Welch correction was performed to determine statistical significance between groups. P values of <0.05 were considered significant.

81

Chapter V

Summary and Future Directions

82

SUMMARY

Utilizing several mouse models, I have identified novel in vivo functions for the

Myocardin Related Transcription Factors as essential regulators of the actin cytoskeleton in brain and muscle development.

I found that conditional deletion of MRTF-A and -B in the mouse brain disrupts neuronal migration and neurite outgrowth, resulting in aberrant development of multiple brain structures. I showed that MRTFs organize the actin cytoskeleton by regulating the expression and the activity of the actin severing proteins Gelsolin and Cofilin. I also identified Pctaire-1 as a novel target gene of the MRTF/SRF pathway that functions in a kinase cascade to regulate actin dynamics, neurite outgrowth and neuronal migration during neurogenesis.

By generating mice lacking both MRTF-A and -B in the heart, I showed that cardiac deletion of MRTFs causes an array of cardiac defects, whose severity correlates with the number of deleted MRTF alleles and that range from reduced cardiac contractility to neonatal lethality with major defects in sarcomere arrangement.

Importantly, I showed that Pctaire-1, whose functions in the heart are completely unknown, acts as a regulator of sarcomere arrangement and an effector of MRTF functions in cardiogenesis.

Moreover, I examined the in vivo functions of MASTR in mice by generating a

MASTR null allele. MASTR knockout mice were born at Mendelian ratios and do not show any major phenotype. However, deletion of MASTR and MRTF-A causes perinatal lethality, possibly due to defective skeletal muscle development.

83

FUTURE DIRECTIONS

These discoveries motivated us to further pursue the molecular mechanisms governing MRTF and Pctaire-1 functions in the heart. As stated earlier, the results of this study are the first to show a role for Pctaire-1 in muscle. We are currently pursuing the analysis of the phenotype of alpha-MHC-Pctaire-1 transgenic mice by examining the progression of the cardiac dilation phenotype with age. Using Pctaire-1 overexpressing mice, we also want to confirm by electron microscopy that Pctaire-1 is indeed a regulator of sarcomere assembly in vivo. In light of this suggested role for Pctaire-1 in cytoskeletal dynamics and sarcomere assembly, and the well studied process of sarcomere and cytoskeletal remodeling in the hypertrophic heart in response to stress, we predict that

Pctaire-1 functions during the cardiac response to hypertrophic stress signals. Therefore, we will subject normal and Pctaire-1 overexpressing hearts to pressure overload by aortic banding to analyze the putative functions of Pctaire-1 in heart pathology in response to stress. At the molecular level, we reported here that the phosphorylation of Cofilin is moderately dysregulated in MRTF cdKO and Pctaire-1 overexpressing hearts, suggesting that the molecular mechanism described in the brain is conserved in the heart as well.

We are currently analyzing the expression and activity of the remaining components of the Pctaire-1/Cdk5 kinase cascade in MRTF cdKO and Pctaire-1 overexpressing hearts.

At the same time, we are performing a yeast-two-hybrid screen in the hope of uncovering novel Pctaire-1 interacting proteins that could link its function to other signaling pathways in the heart.

84

Moreover, our findings that MRTF-A and MASTR cooperate to regulate skeletal muscle development drove us to further pursue the role of MASTR in muscle development. First, we want to confirm that the lethality of MRTF-A/MASTR dKO mice is due exclusively to skeletal muscle defects by ruling out additional possible cardiac or brain defects. We also want to further investigate the molecular mechanisms mediating MASTR activity in muscle cells. As stated earlier, the skeletal muscle defects observed upon deletion of MRTF-A and MASTR suggest that MASTR functions are not exclusively MEF2 dependent. One approach is to examine whether MASTR activates

SRF dependent gene transcription by comparing the expression of SRF target genes in

MRTF-A KO and MRTF-A/MASTR dKO skeletal muscle. Our second approach is to perform a yeast-two-hybrid screen using MASTR as bait in the hope of identifying novel

MASTR partners in muscle cells and uncovering other transcription factors that could be activated by MASTR.

85

Bibliography

Abe, H., Obinata, T., Minamide, L.S., and Bamburg, J.R. (1996). Xenopus laevis actin- depolymerizing factor/cofilin: a phosphorylation-regulated protein essential for development. J Cell Biol 132, 871-885.

Ahlemeyer, B., and Baumgart-Vogt, E. (2005). Optimized protocols for the simultaneous preparation of primary neuronal cultures of the neocortex, hippocampus and cerebellum from individual newborn (P0.5) C57Bl/6J mice. J Neurosci Methods 149, 110-120.

Alberti, S., Krause, S.M., Kretz, O., Philippar, U., Lemberger, T., Casanova, E., Wiebel, F.F., Schwarz, H., Frotscher, M., Schutz, G., et al. (2005). Neuronal migration in the murine rostral migratory stream requires serum response factor. Proc Natl Acad Sci U S A 102, 6148-6153.

Alonso, M., Goodwin, C., Liao, X., Ortiga-Carvalho, T., Machado, D.S., Wondisford, F.E., Refetoff, S., and Weiss, R.E. (2009). In vivo interaction of steroid receptor coactivator (SRC)-1 and the activation function-2 domain of the thyroid hormone receptor (TR) beta in TRbeta E457A knock-in and SRC-1 knockout mice. Endocrinology 150, 3927-3934.

Apostolakis, E.M., Ramamurphy, M., Zhou, D., Onate, S., and O'Malley, B.W. (2002). Acute disruption of select steroid receptor coactivators prevents reproductive behavior in rats and unmasks genetic adaptation in knockout mice. Mol Endocrinol 16, 1511-1523.

Arber, S., Barbayannis, F.A., Hanser, H., Schneider, C., Stanyon, C.A., Bernard, O., and Caroni, P. (1998). Regulation of actin dynamics through phosphorylation of cofilin by LIM-kinase. Nature 393, 805-809.

Arsenian, S., Weinhold, B., Oelgeschlager, M., Ruther, U., and Nordheim, A. (1998). Serum response factor is essential for mesoderm formation during mouse embryogenesis. EMBO J 17, 6289-6299.

Ayala, R., Shu, T., and Tsai, L.H. (2007). Trekking across the brain: the journey of neuronal migration. Cell 128, 29-43.

Bajaj, N.P. (2000). Cyclin-dependent kinase-5 (CDK5) and amyotrophic lateral sclerosis. Amyotroph Lateral Scler Other Motor Neuron Disord 1, 319-327.

Bell, R.D., Deane, R., Chow, N., Long, X., Sagare, A., Singh, I., Streb, J.W., Guo, H., Rubio, A., Van Nostrand, W., et al. (2009). SRF and myocardin regulate LRP-mediated amyloid-beta clearance in brain vascular cells. Nat Cell Biol 11, 143-153.

Bellenchi, G.C., Gurniak, C.B., Perlas, E., Middei, S., Ammassari-Teule, M., and Witke, W. (2007). N-cofilin is associated with neuronal migration disorders and cell cycle control in the cerebral cortex. Genes Dev 21, 2347-2357.

86

Benitez-King, G., Ramirez-Rodriguez, G., Ortiz, L., and Meza, I. (2004). The neuronal cytoskeleton as a potential therapeutical target in neurodegenerative diseases and schizophrenia. Curr Drug Targets CNS Neurol Disord 3, 515-533.

Besset, V., Rhee, K., and Wolgemuth, D.J. (1998). The identification and characterization of expression of Pftaire-1, a novel Cdk family member, suggest its function in the mouse testis and nervous system. Mol Reprod Dev 50, 18-29.

Besset, V., Rhee, K., and Wolgemuth, D.J. (1999). The cellular distribution and kinase activity of the Cdk family member Pctaire1 in the adult mouse brain and testis suggest functions in differentiation. Cell Growth Differ 10, 173-181.

Cao, D., Wang, Z., Zhang, C.L., Oh, J., Xing, W., Li, S., Richardson, J.A., Wang, D.Z., and Olson, E.N. (2005). Modulation of smooth muscle gene expression by association of histone acetyltransferases and deacetylases with myocardin. Mol Cell Biol 25, 364-376.

Cen, B., Selvaraj, A., Burgess, R.C., Hitzler, J.K., Ma, Z., Morris, S.W., and Prywes, R. (2003). Megakaryoblastic leukemia 1, a potent transcriptional coactivator for serum response factor (SRF), is required for serum induction of SRF target genes. Mol Cell Biol 23, 6597-6608.

Charvet, C., Houbron, C., Parlakian, A., Giordani, J., Lahoute, C., Bertrand, A., Sotiropoulos, A., Renou, L., Schmitt, A., Melki, J., et al. (2006). New role for serum response factor in postnatal skeletal muscle growth and regeneration via the interleukin 4 and insulin-like growth factor 1 pathways. Mol Cell Biol 26, 6664-6674.

Chen, F., Dong, J., Wang, F., Wu, F., Zhang, G., Li, G., Chen, Z., Chen, J., and Wei, K. (2007). Identification of barley genotypes with low grain Cd accumulation and its interaction with four microelements. Chemosphere 67, 2082-2088.

Cheng, K., Li, Z., Fu, W.Y., Wang, J.H., Fu, A.K., and Ip, N.Y. (2002). Pctaire1 interacts with p35 and is a novel substrate for Cdk5/p35. J Biol Chem 277, 31988-31993.

Chow, N., Bell, R.D., Deane, R., Streb, J.W., Chen, J., Brooks, A., Van Nostrand, W., Miano, J.M., and Zlokovic, B.V. (2007). Serum response factor and myocardin mediate arterial hypercontractility and cerebral blood flow dysregulation in Alzheimer's phenotype. Proc Natl Acad Sci U S A 104, 823-828.

Creemers, E.E., Sutherland, L.B., Oh, J., Barbosa, A.C., and Olson, E.N. (2006). Coactivation of MEF2 by the SAP domain proteins myocardin and MASTR. Mol Cell 23, 83-96.

Cunningham, C.C., Stossel, T.P., and Kwiatkowski, D.J. (1991). Enhanced motility in NIH 3T3 fibroblasts that overexpress gelsolin. Science 251, 1233-1236.

Dent, E.W., and Gertler, F.B. (2003). Cytoskeletal dynamics and transport in growth cone motility and axon guidance. Neuron 40, 209-227.

87

Descot, A., Rex-Haffner, M., Courtois, G., Bluteau, D., Menssen, A., Mercher, T., Bernard, O.A., Treisman, R., and Posern, G. (2008). OTT-MAL is a deregulated activator of serum response factor-dependent gene expression. Mol Cell Biol 28, 6171-6181.

Dhavan, R., and Tsai, L.H. (2001). A decade of CDK5. Nat Rev Mol Cell Biol 2, 749- 759.

Du, K.L., Chen, M., Li, J., Lepore, J.J., Mericko, P., and Parmacek, M.S. (2004). Megakaryoblastic leukemia factor-1 transduces cytoskeletal signals and induces smooth muscle cell differentiation from undifferentiated embryonic stem cells. J Biol Chem 279, 17578-17586.

Etkin, A., Alarcon, J.M., Weisberg, S.P., Touzani, K., Huang, Y.Y., Nordheim, A., and Kandel, E.R. (2006). A role in learning for SRF: deletion in the adult forebrain disrupts LTD and the formation of an immediate memory of a novel context. Neuron 50, 127-143.

Furnish, E.J., Zhou, W., Cunningham, C.C., Kas, J.A., and Schmidt, C.E. (2001). Gelsolin overexpression enhances neurite outgrowth in PC12 cells. FEBS Lett 508, 282- 286.

Graeser, R., Gannon, J., Poon, R.Y., Dubois, T., Aitken, A., and Hunt, T. (2002). Regulation of the CDK-related protein kinase PCTAIRE-1 and its possible role in neurite outgrowth in Neuro-2A cells. J Cell Sci 115, 3479-3490.

Guettler, S., Vartiainen, M.K., Miralles, F., Larijani, B., and Treisman, R. (2008). RPEL motifs link the serum response factor cofactor MAL but not myocardin to Rho signaling via actin binding. Mol Cell Biol 28, 732-742.

Gurniak, C.B., Perlas, E., and Witke, W. (2005). The actin depolymerizing factor n- cofilin is essential for neural tube morphogenesis and neural crest cell migration. Dev Biol 278, 231-241.

Han, Z., Li, X., Wu, J., and Olson, E.N. (2004). A myocardin-related transcription factor regulates activity of serum response factor in Drosophila. Proc Natl Acad Sci U S A 101, 12567-12572.

Huang, J., Cheng, L., Li, J., Chen, M., Zhou, D., Lu, M.M., Proweller, A., Epstein, J.A., and Parmacek, M.S. (2008). Myocardin regulates expression of contractile genes in smooth muscle cells and is required for closure of the ductus arteriosus in mice. J Clin Invest 118, 515-525.

Iyer, D., Belaguli, N., Fluck, M., Rowan, B.G., Wei, L., Weigel, N.L., Booth, F.W., Epstein, H.F., Schwartz, R.J., and Balasubramanyam, A. (2003). Novel phosphorylation target in the serum response factor MADS box regulates alpha-actin transcription. Biochemistry 42, 7477-7486.

Iyer, D., Chang, D., Marx, J., Wei, L., Olson, E.N., Parmacek, M.S., Balasubramanyam, A., and Schwartz, R.J. (2006). Serum response factor MADS box serine-162

88

phosphorylation switches proliferation and myogenic gene programs. Proc Natl Acad Sci U S A 103, 4516-4521.

Kihara, T., Kano, F., and Murata, M. (2008). Modulation of SRF-dependent gene expression by association of SPT16 with MKL1. Exp Cell Res 314, 629-637.

Kim, Y., Phan, D., van Rooij, E., Wang, D.Z., McAnally, J., Qi, X., Richardson, J.A., Hill, J.A., Bassel-Duby, R., and Olson, E.N. (2008). The MEF2D transcription factor mediates stress-dependent cardiac remodeling in mice. J Clin Invest 118, 124-132.

Knoll, B., Kretz, O., Fiedler, C., Alberti, S., Schutz, G., Frotscher, M., and Nordheim, A. (2006). Serum response factor controls neuronal circuit assembly in the hippocampus. Nat Neurosci 9, 195-204.

Kuhn, T.B., Meberg, P.J., Brown, M.D., Bernstein, B.W., Minamide, L.S., Jensen, J.R., Okada, K., Soda, E.A., and Bamburg, J.R. (2000). Regulating actin dynamics in neuronal growth cones by ADF/cofilin and rho family GTPases. J Neurobiol 44, 126-144.

Kuwahara, K., Barrientos, T., Pipes, G.C., Li, S., and Olson, E.N. (2005). Muscle- specific signaling mechanism that links actin dynamics to serum response factor. Mol Cell Biol 25, 3173-3181.

Lambert de Rouvroit, C., and Goffinet, A.M. (2001). Neuronal migration. Mech Dev 105, 47-56.

Latasa, M.U., Couton, D., Charvet, C., Lafanechere, A., Guidotti, J.E., Li, Z., Tuil, D., Daegelen, D., Mitchell, C., and Gilgenkrantz, H. (2007). Delayed liver regeneration in mice lacking liver serum response factor. Am J Physiol Gastrointest Liver Physiol 292, G996-G1001.

Lazaro, J.B., Kitzmann, M., Poul, M.A., Vandromme, M., Lamb, N.J., and Fernandez, A. (1997). Cyclin dependent kinase 5, cdk5, is a positive regulator of myogenesis in mouse C2 cells. J Cell Sci 110 ( Pt 10), 1251-1260.

Li, J., Zhu, X., Chen, M., Cheng, L., Zhou, D., Lu, M.M., Du, K., Epstein, J.A., and Parmacek, M.S. (2005a). Myocardin-related transcription factor B is required in cardiac neural crest for smooth muscle differentiation and cardiovascular development. Proc Natl Acad Sci U S A 102, 8916-8921.

Li, S., Chang, S., Qi, X., Richardson, J.A., and Olson, E.N. (2006). Requirement of a myocardin-related transcription factor for development of mammary myoepithelial cells. Mol Cell Biol 26, 5797-5808.

Li, S., Czubryt, M.P., McAnally, J., Bassel-Duby, R., Richardson, J.A., Wiebel, F.F., Nordheim, A., and Olson, E.N. (2005b). Requirement for serum response factor for skeletal muscle growth and maturation revealed by tissue-specific gene deletion in mice. Proc Natl Acad Sci U S A 102, 1082-1087.

89

Li, S., Wang, D.Z., Wang, Z., Richardson, J.A., and Olson, E.N. (2003). The serum response factor coactivator myocardin is required for vascular smooth muscle development. Proc Natl Acad Sci U S A 100, 9366-9370.

Lin, L., Lesnick, T.G., Maraganore, D.M., and Isacson, O. (2009). Axon guidance and synaptic maintenance: preclinical markers for neurodegenerative disease and therapeutics. Trends Neurosci 32, 142-149.

Lin, Q., Lu, J., Yanagisawa, H., Webb, R., Lyons, G.E., Richardson, J.A., and Olson, E.N. (1998). Requirement of the MADS-box transcription factor MEF2C for vascular development. Development 125, 4565-4574.

Lin, Q., Schwarz, J., Bucana, C., and Olson, E.N. (1997). Control of mouse cardiac morphogenesis and myogenesis by transcription factor MEF2C. Science 276, 1404-1407.

Liu, N., and Olson, E.N. (2006). Coactivator control of cardiovascular growth and remodeling. Curr Opin Cell Biol 18, 715-722.

Long, X., Bell, R.D., Gerthoffer, W.T., Zlokovic, B.V., and Miano, J.M. (2008). Myocardin is sufficient for a smooth muscle-like contractile phenotype. Arterioscler Thromb Vasc Biol 28, 1505-1510.

McDonald, O.G., Wamhoff, B.R., Hoofnagle, M.H., and Owens, G.K. (2006). Control of SRF binding to CArG box chromatin regulates smooth muscle gene expression in vivo. J Clin Invest 116, 36-48.

McKinsey, T.A., Zhang, C.L., and Olson, E.N. (2002). MEF2: a calcium-dependent regulator of cell division, differentiation and death. Trends Biochem Sci 27, 40-47.

Meadows, S.M., Warkman, A.S., Salanga, M.C., Small, E.M., and Krieg, P.A. (2008). The myocardin-related transcription factor, MASTR, cooperates with MyoD to activate skeletal muscle gene expression. Proc Natl Acad Sci U S A 105, 1545-1550.

Meberg, A., Otterstad, J.E., Froland, G., Lindberg, H., and Sorland, S.J. (2000). Outcome of congenital heart defects--a population-based study. Acta Paediatr 89, 1344-1351.

Meberg, P.J. (2000). Signal-regulated ADF/cofilin activity and growth cone motility. Mol Neurobiol 21, 97-107.

Meberg, P.J., and Bamburg, J.R. (2000). Increase in neurite outgrowth mediated by overexpression of actin depolymerizing factor. J Neurosci 20, 2459-2469.

Medjkane, S., Perez-Sanchez, C., Gaggioli, C., Sahai, E., and Treisman, R. (2009). Myocardin-related transcription factors and SRF are required for cytoskeletal dynamics and experimental metastasis. Nat Cell Biol 11, 257-268.

Miano, J.M. (2003). Serum response factor: toggling between disparate programs of gene expression. J Mol Cell Cardiol 35, 577-593.

90

Miano, J.M., Ramanan, N., Georger, M.A., de Mesy Bentley, K.L., Emerson, R.L., Balza, R.O., Jr., Xiao, Q., Weiler, H., Ginty, D.D., and Misra, R.P. (2004). Restricted inactivation of serum response factor to the cardiovascular system. Proc Natl Acad Sci U S A 101, 17132-17137.

Miralles, F., Posern, G., Zaromytidou, A.I., and Treisman, R. (2003). Actin dynamics control SRF activity by regulation of its coactivator MAL. Cell 113, 329-342.

Mouilleron, S., Guettler, S., Langer, C.A., Treisman, R., and McDonald, N.Q. (2008). Molecular basis for G-actin binding to RPEL motifs from the serum response factor coactivator MAL. EMBO J 27, 3198-3208.

Naya, F.J., Black, B.L., Wu, H., Bassel-Duby, R., Richardson, J.A., Hill, J.A., and Olson, E.N. (2002). Mitochondrial deficiency and cardiac sudden death in mice lacking the MEF2A transcription factor. Nat Med 8, 1303-1309.

Nishihara, E., Yoshida-Komiya, H., Chan, C.S., Liao, L., Davis, R.L., O'Malley, B.W., and Xu, J. (2003). SRC-1 null mice exhibit moderate motor dysfunction and delayed development of cerebellar Purkinje cells. J Neurosci 23, 213-222.

Niu, Z., Yu, W., Zhang, S.X., Barron, M., Belaguli, N.S., Schneider, M.D., Parmacek, M., Nordheim, A., and Schwartz, R.J. (2005). Conditional mutagenesis of the murine serum response factor gene blocks cardiogenesis and the transcription of downstream gene targets. J Biol Chem 280, 32531-32538.

Oh, J., Richardson, J.A., and Olson, E.N. (2005). Requirement of myocardin-related transcription factor-B for remodeling of branchial arch arteries and smooth muscle differentiation. Proc Natl Acad Sci U S A 102, 15122-15127.

Okuda, T., Cleveland, J.L., and Downing, J.R. (1992). PCTAIRE-1 and PCTAIRE-3, two members of a novel cdc2/CDC28-related protein kinase gene family. Oncogene 7, 2249- 2258.

Park, H.T., Wu, J., and Rao, Y. (2002). Molecular control of neuronal migration. Bioessays 24, 821-827.

Parlakian, A., Charvet, C., Escoubet, B., Mericskay, M., Molkentin, J.D., Gary-Bobo, G., De Windt, L.J., Ludosky, M.A., Paulin, D., Daegelen, D., et al. (2005). Temporally controlled onset of dilated cardiomyopathy through disruption of the SRF gene in adult heart. Circulation 112, 2930-2939.

Parlakian, A., Tuil, D., Hamard, G., Tavernier, G., Hentzen, D., Concordet, J.P., Paulin, D., Li, Z., and Daegelen, D. (2004). Targeted inactivation of serum response factor in the developing heart results in myocardial defects and embryonic lethality. Mol Cell Biol 24, 5281-5289.

Parmacek, M.S. (2007). Myocardin-related transcription factors: critical coactivators regulating cardiovascular development and adaptation. Circ Res 100, 633-644.

91

Philpott, A., Porro, E.B., Kirschner, M.W., and Tsai, L.H. (1997). The role of cyclin- dependent kinase 5 and a novel regulatory subunit in regulating muscle differentiation and patterning. Genes Dev 11, 1409-1421.

Pipes, G.C., Creemers, E.E., and Olson, E.N. (2006). The myocardin family of transcriptional coactivators: versatile regulators of cell growth, migration, and myogenesis. Genes Dev 20, 1545-1556.

Posern, G., Miralles, F., Guettler, S., and Treisman, R. (2004). Mutant actins that stabilise F-actin use distinct mechanisms to activate the SRF coactivator MAL. EMBO J 23, 3973- 3983.

Posern, G., and Treisman, R. (2006). Actin' together: serum response factor, its cofactors and the link to signal transduction. Trends Cell Biol 16, 588-596.

Potthoff, M.J., Arnold, M.A., McAnally, J., Richardson, J.A., Bassel-Duby, R., and Olson, E.N. (2007a). Regulation of skeletal muscle sarcomere integrity and postnatal muscle function by Mef2c. Mol Cell Biol 27, 8143-8151.

Potthoff, M.J., and Olson, E.N. (2007). MEF2: a central regulator of diverse developmental programs. Development 134, 4131-4140.

Potthoff, M.J., Wu, H., Arnold, M.A., Shelton, J.M., Backs, J., McAnally, J., Richardson, J.A., Bassel-Duby, R., and Olson, E.N. (2007b). Histone deacetylase degradation and MEF2 activation promote the formation of slow-twitch myofibers. J Clin Invest 117, 2459-2467.

Ramanan, N., Shen, Y., Sarsfield, S., Lemberger, T., Schutz, G., Linden, D.J., and Ginty, D.D. (2005). SRF mediates activity-induced gene expression and synaptic plasticity but not neuronal viability. Nat Neurosci 8, 759-767.

Rossi, F., Gianola, S., and Corvetti, L. (2007). Regulation of intrinsic neuronal properties for axon growth and regeneration. Prog Neurobiol 81, 1-28.

Sarmiere, P.D., and Bamburg, J.R. (2004). Regulation of the neuronal actin cytoskeleton by ADF/cofilin. J Neurobiol 58, 103-117.

Sawada, T., Nishiyama, C., Kishi, T., Sasazuki, T., Komazawa-Sakon, S., Xue, X., Piao, J.H., Ogata, H., Nakayama, J., Taki, T., et al. (2008). Fusion of OTT to BSAC results in aberrant up-regulation of transcriptional activity. J Biol Chem 283, 26820-26828.

Selvaraj, A., and Prywes, R. (2003). Megakaryoblastic leukemia-1/2, a transcriptional co- activator of serum response factor, is required for skeletal myogenic differentiation. J Biol Chem 278, 41977-41987.

Shelton, J.M., Lee, M.H., Richardson, J.A., and Patel, S.B. (2000). Microsomal triglyceride transfer protein expression during mouse development. J Lipid Res 41, 532- 537.

92

Shiota, J., Ishikawa, M., Sakagami, H., Tsuda, M., Baraban, J.M., and Tabuchi, A. (2006). Developmental expression of the SRF co-activator MAL in brain: role in regulating dendritic morphology. J Neurochem 98, 1778-1788.

Small, E.M., Warkman, A.S., Wang, D.Z., Sutherland, L.B., Olson, E.N., and Krieg, P.A. (2005). Myocardin is sufficient and necessary for cardiac gene expression in Xenopus. Development 132, 987-997.

Smith, D. (2003). Cdk5 in neuroskeletal dynamics. Neurosignals 12, 239-251.

Spalice, A., Parisi, P., Nicita, F., Pizzardi, G., Del Balzo, F., and Iannetti, P. (2009). Neuronal migration disorders: clinical, neuroradiologic and genetics aspects. Acta Paediatr 98, 421-433.

Spiegelman, B.M., and Heinrich, R. (2004). Biological control through regulated transcriptional coactivators. Cell 119, 157-167.

Stern, S., Debre, E., Stritt, C., Berger, J., Posern, G., and Knoll, B. (2009). A nuclear actin function regulates neuronal motility by serum response factor-dependent gene transcription. J Neurosci 29, 4512-4518.

Sun, Q., Chen, G., Streb, J.W., Long, X., Yang, Y., Stoeckert, C.J., Jr., and Miano, J.M. (2006). Defining the mammalian CArGome. Genome Res 16, 197-207.

Tanaka, D.H., Yamauchi, K., and Murakami, F. (2008). [Guidance mechanisms in neuronal and axonal migration]. Brain Nerve 60, 405-413.

Tevosian, S.G., Deconinck, A.E., Tanaka, M., Schinke, M., Litovsky, S.H., Izumo, S., Fujiwara, Y., and Orkin, S.H. (2000). FOG-2, a cofactor for GATA transcription factors, is essential for heart morphogenesis and development of coronary vessels from epicardium. Cell 101, 729-739.

Torrado, M., Lopez, E., Centeno, A., Medrano, C., Castro-Beiras, A., and Mikhailov, A.T. (2003). Myocardin mRNA is augmented in the failing myocardium: expression profiling in the porcine model and human dilated cardiomyopathy. J Mol Med 81, 566-577.

Verrotti, A., Spalice, A., Ursitti, F., Papetti, L., Mariani, R., Castronovo, A., Mastrangelo, M., and Iannetti, P. (2009). New trends in neuronal migration disorders. Eur J Paediatr Neurol.

Wang, D., Chang, P.S., Wang, Z., Sutherland, L., Richardson, J.A., Small, E., Krieg, P.A., and Olson, E.N. (2001). Activation of cardiac gene expression by myocardin, a transcriptional cofactor for serum response factor. Cell 105, 851-862.

Wang, D.Z., Li, S., Hockemeyer, D., Sutherland, L., Wang, Z., Schratt, G., Richardson, J.A., Nordheim, A., and Olson, E.N. (2002). Potentiation of serum response factor activity by a family of myocardin-related transcription factors. Proc Natl Acad Sci U S A 99, 14855-14860.

93

Wang, Z., Wang, D.Z., Hockemeyer, D., McAnally, J., Nordheim, A., and Olson, E.N. (2004). Myocardin and ternary complex factors compete for SRF to control smooth muscle gene expression. Nature 428, 185-189.

Witke, W., Sharpe, A.H., Hartwig, J.H., Azuma, T., Stossel, T.P., and Kwiatkowski, D.J. (1995). Hemostatic, inflammatory, and fibroblast responses are blunted in mice lacking gelsolin. Cell 81, 41-51.

Xing, W., Zhang, T.C., Cao, D., Wang, Z., Antos, C.L., Li, S., Wang, Y., Olson, E.N., and Wang, D.Z. (2006). Myocardin induces cardiomyocyte hypertrophy. Circ Res 98, 1089-1097.

Yang, N., Higuchi, O., Ohashi, K., Nagata, K., Wada, A., Kangawa, K., Nishida, E., and Mizuno, K. (1998). Cofilin phosphorylation by LIM-kinase 1 and its role in Rac- mediated actin reorganization. Nature 393, 809-812.

Zaromytidou, A.I., Miralles, F., and Treisman, R. (2006). MAL and ternary complex factor use different mechanisms to contact a common surface on the serum response factor DNA-binding domain. Mol Cell Biol 26, 4134-4148.

Zhang, Z., Ottens, A.K., Larner, S.F., Kobeissy, F.H., Williams, M.L., Hayes, R.L., and Wang, K.K. (2006). Direct Rho-associated kinase inhibiton induces cofilin dephosphorylation and neurite outgrowth in PC-12 cells. Cell Mol Biol Lett 11, 12-29.

Zhuo, L., Theis, M., Alvarez-Maya, I., Brenner, M., Willecke, K., and Messing, A. (2001). hGFAP-cre transgenic mice for manipulation of glial and neuronal function in vivo. Genesis 31, 85-94.

94

71