+ All Categories
Home > Documents > By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf ·...

By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf ·...

Date post: 28-Feb-2019
Category:
Upload: hadieu
View: 213 times
Download: 0 times
Share this document with a friend
121
THE MECHANICS OF NUCLEAR SHAPING IN CELL By YUAN LI A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF FLORIDA IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY UNIVERSITY OF FLORIDA 2017
Transcript
Page 1: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

THE MECHANICS OF NUCLEAR SHAPING IN CELL

By

YUAN LI

A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF FLORIDA IN PARTIAL FULFILLMENT

OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY

UNIVERSITY OF FLORIDA

2017

Page 2: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

© 2017 Yuan Li

Page 3: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

To my beloved parents

Page 4: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

4

ACKNOWLEDGMENTS

I would like to acknowledge and thank the people who supported me during this

crucial time of my life. The first and most important one is my advisor, Dr. Tanmay Lele,

who provided me guidance and support with great patience and attention during my

whole time in the lab. He helped me to improve my logical and critical thinking as an

independent researcher. His interesting ideas inspired me to build my original idea and

maintained my motivation on research projects. He did his best to offer a healthy and

collaborative environment to perform research. I will always be grateful to him.

I would like to thank my co-advisor Dr. Richard Dickinson who gave a lot of

helpful advices and comments on the research projects. He was always willing to meet

and discuss with me regarding the research questions, and his expertise in

mathematics modeling helped a lot on my project. I would also like to thank my other

committee members Dr. Alexander Ishov and Dr. Rinaldi Carlos for their support on

experiment and encouraging words. I would like to thank Dr. Gregg Gundersen, Dr.

Jeffery Nickerson and Dr. Kyle Roux with whom I collaborated on different projects.

They always responded to my questions timely with their expertise and provided

important plasmids and engineered cell lines that promoted my research.

I would like to thank my former lab mates Dr. Jun Wu, Dr. TJ chancellor and Dr.

David Lovett who trained me on all basic techniques in the lab and helped me design

experiments in the beginning of my research. I would like to thank my former and

current lab mates Dr. Srujana Neelem, Dr. Samer Alam, Dr. Ian Kent and Dr. Varun

Agrawal, Qiao Zhang, VJ Tocco, Keith Christopher and Andrew Tamashunas, with

whom I spent almost my lab time. The valuable discussions and interesting chats with

them gave me a helpful and fun lab environment to work in. I would also like to thank

Page 5: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

5

Dr. Jun Yin and Dr. Shen-Hsiu Hung from Dr. Yiider Tseng’s Lab for providing their

experienced ideas and experiment facilities. I would also like to thank the former master

students Nandini Shekhar, Agnes Mendonca and Anirudh Ram and undergraduate

students Catherine Perez, Alaina Giacobbe, Aniruddha Shirhatti and Uday Rallabhandi

who worked with me on various projects. I would like to thank all my UF friends who

encouraged and helped me to go through this adventure of research.

Finally, I would like to thank my family members who are my strongest support to

achieve my goals. My mother Yinchi Liu always encouraged me to bravely fight with

difficulties in life and to never give up my dreams. Her support and sacrifice throughout

these years made my dream come true. My father Heng Li taught me to keep a

peaceful mind no matter what happens around you. It helped me survive from the two

hard times throughout this pursuing journey. I would also like to thank my other family

members, especially my beloved grandparents who made me feel loved and were

always proud of me.

Page 6: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

6

TABLE OF CONTENTS page

ACKNOWLEDGMENTS .................................................................................................. 4

LIST OF TABLES ............................................................................................................ 8

LIST OF FIGURES .......................................................................................................... 9

LIST OF ABBREVIATIONS ........................................................................................... 11

ABSTRACT ................................................................................................................... 13

CHAPTER

1 INTRODUCTION .................................................................................................... 15

2 MOVING CELL BOUNDARY DRIVES NUCLEAR FLATTENING DURING CELL SPREADING ........................................................................................................... 21

Materials and Methods............................................................................................ 22 Cell Culture, Plasmids and Drug Treatment ..................................................... 22

Cell Spreading and Trysinization Assay ........................................................... 22 Fixation and Immunocytochemistry .................................................................. 23

Protein Silencing .............................................................................................. 24

Western Blotting ............................................................................................... 24

Microscopy and Image Analysis ....................................................................... 25 Results .................................................................................................................... 25

Collapse of Apical Nuclear Surface Contributes to Nuclear Flattening during Early Cell Spreading ..................................................................................... 25

Nuclear Flattening does Not Require Actomyosin Contraction in Spreading Cells .............................................................................................................. 27

Intermediate Filaments and Microtubules are Dispensable for Nuclear Flattening in Spreading Cells ........................................................................ 29

Apical and Basal Actomyosin Bundles are Not Required for Nuclear Flattening during Initial Cell Spreading .......................................................... 30

Nuclear Flattening can be Reversed by Detachment of the Cell from the Substratum .................................................................................................... 31

The LINC Complex is Not Required for Nuclear Flattening .............................. 32 A Mathematical Model for Nuclear Flattening and Cell Spreading ................... 33

Discussion .............................................................................................................. 39

3 DYNAMIC DEFORMATION OF THE CELL PLASTICALLY SHAPES THE NUCLEUS AND AMPLIFIES CANCER NUCLEAR IRREGULARITIES.................. 71

Materials and Methods............................................................................................ 72

Cell Culture and Transfection ........................................................................... 72

Page 7: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

7

Cell Staining and Drug Treatment .................................................................... 73 Nuclear Excision ............................................................................................... 74

Cell Spreading Assay ....................................................................................... 74 Imaging and Image Analysis ............................................................................ 74 High-Content Imaging ...................................................................................... 75

Results .................................................................................................................... 75 The Deforming Cell Shape Amplifies Cancer Nuclear Abnormalities ............... 75

Disrupting Either the LINC Complex or the Cytoskeleton Dampens Cancer Nuclear Abnormality ...................................................................................... 76

The Reduction of Nuclear Abnormality by LINC Complex Disruption Impairs Cellular Motility .............................................................................................. 77

Abnormal Morphology of Cancer Nucleus do NOT Necessarily Reflect Chromatin Content ........................................................................................ 77

Nuclear Abnormality can be Inherited by Offspring .......................................... 78 Discussion .............................................................................................................. 79

Nuclear Abnormalities in Cancer. ..................................................................... 79

Mechanical Stress and Cancer Cell Migration. ................................................. 81

4 CONCLUSIONS ..................................................................................................... 92

Summary of Findings .............................................................................................. 92

Future Work ............................................................................................................ 94

APPENDIX

A COMPUTATIONAL MODEL FOR NUCLEAR DEFORMATION DURING CELL SPREADING ........................................................................................................... 99

Constitutive Model for Cytoskeletal Network Stress ......................................... 99

Model for Cell Mechanics ............................................................................... 101 Model Parameters .......................................................................................... 103

Methods for Simulating Cell Spreading .......................................................... 105

B THE INFLUENCE OF CELL GEOMETRY ON NUCLEAR VOLUME ................... 108

LIST OF REFERENCES ............................................................................................. 111

BIOGRAPHICAL SKETCH .......................................................................................... 121

Page 8: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

8

LIST OF TABLES

Table page A-1 Parameters of Cell Spreading Model ................................................................ 107

Page 9: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

9

LIST OF FIGURES

Figure page 1-1 Representative IF images of nuclear morphology .............................................. 19

1-2 Illustrating cartoon of LINC complex. .................................................................. 20

2-1 The dynamics of nuclear flattening during early cell spreading. ......................... 43

2-2 The nuclear deformation causes nuclear flattening but not already-flat nucleus toppling onto their side .......................................................................... 44

2-3 The dynamics of nuclear flattening against substratum is not influenced by gravity ................................................................................................................. 45

2-4 Nuclei does not flatten when cell spreading is prevented by inhibitors of actin assembly ............................................................................................................ 46

2-5 The nucleus flattens completely in a partially spread cell ................................... 47

2-6 The apical surface of cell was separated from the apical surface of nucleus in the early stage of cell spreading ......................................................................... 48

2-7 Nuclear flattening is independent of actomyosin contraction .............................. 49

2-8 The effect of myosin inhibition on nuclear shape in well spread cell ................... 51

2-9 Inhibition of myosin activity with Y-27632 does not alter the qualitative features of dynamic nuclear flattening during cell spreading ............................ 53

2-10 Blebbistatin prevents cell spreading at later times resulting in final nuclear rounding ............................................................................................................. 54

2-11 The absence of intermediate filaments does not prevent nuclear flattening during cell spreading .......................................................................................... 55

2-12 Disruption of microtubules by nocodazole rounds up the nucleus but also prevents cell spreading ....................................................................................... 56

2-13 The absence of microtubule does not prevent nuclear flattening during cell spreading ............................................................................................................ 57

2-14 Apical and basal actomyosin bundles are not required for nuclear flattening during initial cell spreading ................................................................................. 58

2-15 Nuclear flattening can be reversed by detachment of the cell from the substratum. ......................................................................................................... 60

Page 10: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

10

2-16 The LINC complex is not required for nuclear flattening during cell spreading ... 61

2-17 GFP-KASH4 overexpression slows down but does not prevent cell spreading and nuclear flattening ......................................................................................... 63

2-18 Nuclei in lamin A/C -/- MEFs flatten faster than WT cells ................................... 65

2-19 Mathematical model for nuclear deformation during cell spreading .................... 67

2-20 Snapshots of simulation results for different parameters .................................... 69

3-1 The abnormality of nucleus amplifies during cell spreading ............................... 83

3-2 Disrupting the LINC complex reduces nuclear abnormality ................................ 85

3-3 Disrupting cytoskeletal elements reduces nuclear abnormality. ......................... 86

3-4 Trajectory maps of MDA-MB-231 cells with or without LINC disruption .............. 87

3-5 LINC complex disruption impairs cellular motility ................................................ 88

3-6 The abnormal shape of nucleus does not correlate with DNA content. .............. 89

3-7 The abnormality of nuclear shape is heritable .................................................... 91

4-1 Data pool of x-z nuclear aspect ratio versus cell spreading area........................ 97

4-2 Local nuclear deformation in response to local protrusion and retraction of cell memrbane .................................................................................................... 98

B-1 The difference of nuclear volume induced by cell geometry vanishes after the removal of geometry constrain ......................................................................... 109

Page 11: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

11

LIST OF ABBREVIATIONS

3T3 3-day Transfer, Inoculum 3x105 Cells, a mouse embryonic fibroblast cell Line

Blebb Blebbistatin

Colc Colcemid

Crtl Control

Cyto D Cytochalasin D

DBS Donor Bovine Serum

DIC Differential Interference Contrast

DMEM Dulbecco’s Modified Eagle’s Medium

EGF Epidermal Growth Factor

EGFP Enhanced Green Fluorescent Protein

FN Fibronectin

FRAP Fluorescence Recovery After Photo-bleaching

GFP Green Fluorescent Protein

IF Intermediate Filament

INM Inner Nuclear Membrane

KASH Klarsicht/Anc-1/Syne Homology

KD knockdown

KDEL Lysine/Aspartic acid/Glutamic acid/Leucine

LAP Lamin Associated Protein

Lat A/B Latrunculin A/B

LINC Linker of nucleoskeleton to cytoskeleton

MEF Mouse embryonic fibroblast

MEM Modified Eagle’s Medium

Page 12: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

12

mRNA Messenger RNA

MSD Mean Square Displacement

MT Microtubule

n Sample number

N2G Nesprin 2 giant

NA Numerical aperture

NE Nuclear envelope

NIH National Institutes of Health

nM Nanomolar

Noco Nocodazole

ONM Outer Nuclear Membrane

p p-value of statistic test

RFP Red Fluorescent Protein

SEM Standard Error of Mean

shRNA Small hairpin RNA

STD Standard deviation

SUN Sad1p, UNC-84

TAN Transmembrane Actin-associated Nuclear

Vim Vimentin

w/v Weight per volume

WT Wild type

Y27 Y27632

μM Micro molar

Page 13: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

13

Abstract of Dissertation Presented to the Graduate School of the University of Florida in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy

THE MECHANICS OF NUCLEAR SHAPING IN CELL

By

Yuan Li

August 2017

Chair: Tanmay Lele Major: Chemical Engineering

The nucleus has a smooth, regular appearance in normal cells, and its shape is

greatly altered in human pathologies. Yet, how the cell establishes nuclear shape is not

well understood. We imaged the dynamics of nuclear shaping in NIH3T3 fibroblasts.

Nuclei translated toward the substratum and began flattening during the early stages of

cell spreading. Initially, nuclear height and width correlated with the degree of cell

spreading, but over time, reached steady-state values even as the cell continued to

spread. Actomyosin activity, actomyosin bundles, microtubules, and intermediate

filaments, as well as the LINC complex, were all dispensable for nuclear flattening as

long as the cell could spread. Together, these results show that cell spreading is

necessary and sufficient to drive nuclear flattening under a wide range of conditions,

including in the presence or absence of myosin activity. To explain this observation, we

propose a computational model for nuclear and cell mechanics that shows how frictional

transmission of stress from the moving cell boundaries to the nuclear surface shapes

the nucleus during early cell spreading. Our results point to a surprisingly simple

mechanical system in cells for establishing nuclear shapes.

Page 14: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

14

Consistent with the above results, researchers in the Lele lab found that

deformed shapes of nuclei are unchanged even after removal of the cell with micro-

dissection, both for smooth, elongated nuclei in fibroblasts and abnormally shaped

nuclei in breast cancer cells. The lack of shape relaxation implies that the nuclear shape

in spread cells does not store any elastic energy, and the cellular stresses that deform

the nucleus are dissipative, not static. Building on these results, we show that during

cell spreading, the deviation of the nucleus from a convex shape increases in MDA-MB-

231 cancer cells, but decreases in MCF-10A cells. Cancer nuclear abnormalities are

uncorrelated with the amount of DNA in cells. We propose that motion of cell

boundaries exert a stress on the cancer nucleus and this amplifies nuclear

abnormalities. Finally, we report the novel finding that disrupting the LINC complex,

which physically links the nucleus to the cytoskeleton, normalizes cancer nuclear shape

and decreases cancer cell migration.

Page 15: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

15

CHAPTER 1 INTRODUCTION

The nucleus was described and characterized back in the 1800s [1]. Typically,

the nucleus has a smooth elliptical appearance in healthy cells. Nuclear shape becomes

altered in a number of diseases like cancers [2-4] and laminopathies [5, 6] (Figure 1-1).

In pathology, the correlation between changed morphology of nuclei and certain

diseases has been observed for a long time [7]. Yet, the mechanism by which cell

establishes the shape of nucleus, and how the nuclear shape becomes abnormal in

pathologies has remained poorly understood.

The nuclear lamina, a complex structure of A- and B-type lamins [8] and lamin-

associated proteins (LAPs) [9], is a key mechanical shell that surrounds chromatin and

is necessary for stable nuclear shape [10, 11]. Chromatin is anchored to the nuclear

lamina either directly [12] or indirectly through binding with LAPs [13]. Changes to the

shape of the nucleus as observed in pathologies typically are accompanied by down-

regulation of nuclear lamins [14-16]. Such changes can alter the spatial configuration of

chromatin and consequently modulate gene expression [17-19]. Access of chromatin to

transcription factors may be influenced by nuclear shape which can also change gene

expression [20]. Thus, alterations of nuclear shape might themselves be contributors to

the progression of pathologies like cancer.

Cytoskeletal forces are exerted on the nucleus and can cause changes in

nuclear shape and position. The cytoskeletal network consists of intertwining filaments

and tubules throughout the cell [21]. Microfilaments or F-actin filaments are the smallest

components of the cytoskeleton with a diameter of 7 nm. Interacting with myosin II

motor proteins and cross linker proteins, actin filaments form a structure named the

Page 16: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

16

actomyosin cytoskeleton, which generates contractile force in cells. Compression driven

by contraction of actomyosin bundles overlaying the surface of the nucleus has been

proposed to establish the flattened nuclear shapes [22] commonly observed in cell

culture. As we will show in this thesis however, such bundles are absent during nuclear

flattening, and myosin activity is not required for nuclear flattening. Other studies have

proposed that actomyosin exerts tensile force on the nucleus to modulate the nuclear

shape [23, 24]. Actin retrograde flow can also exert shear forces that move the nucleus

[25, 26].

Microtubules are tubular structures with an outer diameter of 25 nm, which

enable motor proteins to walk on and transport molecules throughout the cytoplasm.

Two motor proteins mainly interact with microtubules. Dynein walks toward the minus (-)

end of microtubules; and kinesin walks towards plus (+) end. Both these proteins bind to

the nesprin family proteins [27, 28] which are embedded in the outer nuclear

membrane. In this way, microtubule motors can exert force on the nuclear surface

which has been shown to move it as well as rotate it in [29, 30]. We show in this thesis

that microtubules are dispensable for nuclear flattening, contradicting previous reports

[31].

Cytoplasmic intermediate filaments (IFs) are stable rod-like fibers made of

vimentin in fibroblasts with a diameter of 10 nm. IFs are unable to generate intercellular

force; however, IFs can transmit force indirectly form actomyosin to the nuclear

membrane by interacting with actin filaments [32]. Also, IF networks can wrap around

the nucleus and resist any changes in nuclear shape passively [33]. We show that

intermediate filaments are not required for nuclear flattening.

Page 17: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

17

The so-called LINC (Linker of Nucleoskeleton and Cytoskeleton) complex

transmits mechanical stresses to the nuclear surface. The LINC complex mainly

includes outer nuclear membrane nesprin proteins and inner nuclear membrane SUN

proteins (Figure 1-2). KASH domains of the nesprin proteins bind to SUN trimers in the

perinuclear space, while the extra-nuclear domains of nesprins bind to cytoskeletal

elements. On the nucleoplasmic side, SUN proteins bind to the nuclear lamina and thus

complete the linkage between nucleus and the cytoskeleton [34-36]. Disrupting the

LINC complex disrupts force transmission between the cytoskeleton and nucleus [23,

37]. Therefore, it is possible that the LINC complex transmits mechanical stresses that

shape the nucleus, and others have argued that this is the case [38]. However, here we

show that disrupting the LINC complex slows down the process of cell spreading and

nuclear flattening; presumably because stresses are not efficiently transmitted to the

nuclear surface but does not prevent nuclear flattening.

Overall, in this thesis, we addressed two broad questions: 1) how is the nucleus

shaped in the cell and 2) how does the nucleus become abnormally shaped in cancer?

In chapter 2, we report the discovery of a novel mechanism for nuclear shaping that

contradicts previously explanations proposed in the field. We show that in fibroblasts,

cell spreading is necessary and sufficient to drive nuclear flattening under a wide range

of conditions. We propose a model in which frictional transmission of stress from the

moving cell boundaries to the nuclear surface shapes the nucleus during early cell

spreading. In chapter 3, we report that abnormalities in cancer nuclear shape are

amplified by stresses generated by the motion of cell boundaries during spreading, and

that disruption of the LINC complex and cytoskeleton-based forces reduce nuclear

Page 18: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

18

shape abnormalities. We also show that LINC disruption reduces cancer cell motility.

Collectively, we have proposed a new mechanism for how the nucleus shapes the cell,

and how it becomes abnormal in pathologies.

Page 19: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

19

Figure 1-1. Representative IF images of nuclear morphology in breast cancer cell line

(MDA-MB-231) compared with normal breast cell line (MCF-10A) and Hutchinson-Gilford progeria syndrome (HGPS) cell compared with normal cell [39]; fluorescence label is laminA and laminA/C, respectively.

Page 20: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

20

Figure 1-2. Illustrating cartoon of LINC complex. Main components of LINC complex

are denoted in the picture with corresponding color of the structure.

Page 21: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

21

CHAPTER 2 MOVING CELL BOUNDARY DRIVES NUCLEAR FLATTENING DURING CELL

SPREADING

The nucleus, the largest organelle in mammalian cells, has a smooth, regular

appearance in normal cells. However, nuclear shape becomes altered in a few

pathologies such as cancer [40-44] and in laminopathies [15, 39, 45, 46]. The control of

nuclear shapes for cells is particularly important because nuclear shape may directly

control gene expression [17-19]. How the cell shapes the nucleus is not understood.

Given the high rigidity of the nucleus, significant and dynamic changes in nuclear

shape are expected to require forces that far exceed thermal forces in the cell. Such

forces likely originate in the cytoskeleton, which is known to link to the nuclear surface

through the LINC complex (for linker of nucleoskeleton to cytoskeleton) [34, 47, 48].

Candidates for shaping the nucleus include microtubule motors which can shear the

nuclear surface [29, 30] and intermediate filaments that can passively resist nuclear

shape changes by packing around the nuclear envelope or transmitting forces from

actomyosin contraction to the nuclear surface [33, 49]. The actomyosin cytoskeleton

which can push [22], pull [23, 24] or shear and drag the nuclear surface [25, 26] is also

assumed to be a significant component of the nuclear shaping machinery in the cell.

Here, using a combination of experiments to disrupt the cytoskeleton and the

LINC complex, and mathematical modeling and computation, we show that the motion

of cell boundaries drives changes in nuclear shapes during cell spreading. Our results

Reprinted from Biophysical Journal, volume 109, Li, Y., Lovett, D., Zhang, Q., Neelam, S., Kuchibhotla, R.A., Zhu, R.J., Gundersen, G.G., Lele, T.P., and Dickinson, R.B., Moving Cell Boundaries Drive Nuclear Shaping during Cell Spreading, pages 670-686, Copyright (2015), with permission from Elsevier.

Page 22: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

22

point to a surprisingly simple mechanical system in cells for establishing nuclear

shapes.

Materials and Methods

Cell Culture, Plasmids and Drug Treatment

NIH 3T3 fibroblasts were cultured in Dulbecco’s modified Eagle’s medium

(DMEM) with 4.5g/L glucose (Mediatech, Manassas, VA) supplemented with 10% donor

bovine serum (DBS, Gibco, Grand Island, NY) and 1% Penicillin Streptomycin

(Mediatech, Manassas, VA). MEFs were cultured in DMEM, supplemented with 10%

DBS. All cells were maintained at 37°C in a humidified 5% CO2 environment with

passage at 80% confluence. For microscopy, cells were transferred onto 35 mm glass-

bottom dishes (World precision Instrument, Sarasota, FL) treated with 5 µg/ml

fibronectin (BD Biosciences) at 10% confluence. Transient transfection of plasmids into

cells was performed with Lipofectamine 2000 reagent (Life Technologies/Invitrogen,

Carlsbad, CA) in OPTI-MEM media (Life Technologies/Invitrogen, Carlsbad, CA). For

drug treatment studies, Y-27632 (EMD Millipore, Billerica, MA), Blebbistatin (EMD

Millipore, Billerica, MA) or ML-7 (Sigma-Aldrich) was added to the cells for inhibiting

myosin activity at concentrations of 25 µM, 50 µM and 25 µM, respectively. Nocodazole

or Colcemid (Sigma-Aldrich) was used to disrupt microtubules at a concentration of 0.83

µM and 0.27 µM, respectively. To disrupt F-actin polymerization, cells were treated with

2 µM Cytochalasin D (Biomol, Plymouth Meeting, Pennsylvania) and 5 µM Latrunculin A

(Cayman Chemical, Ann Arbor, Michigan).

Cell Spreading and Trysinization Assay

In the cell spreading assay, cells were trypsinized and then seeded onto

fibronectin coated glass-bottomed dishes. They were next incubated at 37°C in 5% CO2

Page 23: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

23

for varying times and then fixed with 4% paraformaldehyde for 20 minutes. For myosin

inhibition and disruption of F-actin and microtubule, the cells were pre-treated with the

appropriate dose of drug for 1 hour, trypsinized and re-suspended in cell culture

medium containing the same dose of drug. They were next seeded in the presence of

the drug for varying time before fixation. In other drug treatment experiments, cells were

allowed to grow on FN coated glass-bottom dishes for 24 hours. They were then

incubated with drug-containing medium for 1 or 2 hours, after which the cells were fixed

and stained. For cell spreading with inverted coverslips, 18mm coverslips were coated

with FN overnight in first day and next day cells were trypsinized and seeded on the

coverslips for 5min to attach with the substratum. Then the coverslips were put into 12-

wells upside down with tweezer very carefully and stayed in the dish for certain time (for

example, 10min, 30min etc.) before fixing and staining. In the trysinization assay, cells

were transfected with GFP-histone H1.1 and RFP-LifeAct (Ibidi, Verona, WI), and

cultured on fibronectin-coated glass-bottom dishes for 24 hours. After placing the dish

onto the microscope stage, the culture medium was removed and the dish was washed

once very gently with phosphate buffered saline (PBS). Then 0.25% (w/v) trypsin (high

concentration) or 0.08% (w/v) trypsin in serum-free medium (low concentration) were

added to detach the cells from the substratum.

Fixation and Immunocytochemistry

Cells were first fixed with 4% (m/v) paraformaldehyde (Electron Microscopy

Sciences, Hatfield, PA) for 20 minutes, and then mounted with ProLong Gold Antifade

Mountant (Life Technologies). To visualize F-actin and nuclei, the fixed cells were

incubated with 1:40 Alexa Fluor 488 phalloidin (Invitrogen) and 1:100 Hoechst 33342

(Life Technologies) for 1 hour at room temperature, respectively. To immune-stain

Page 24: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

24

microtubules, cells were first treated with microtubule extraction buffer containing 0.5%

(m/v) glutaraldehyde, 0.8% formaldehyde and 0.5% Triton X-100 in phosphate buffered

saline (PBS) for 3 minutes before fixing with 1% (m/v) paraformaldehyde for another 10

minutes. Then a freshly prepared 1% (w/v) sodium borohydride in PBS solution was

added to the cells for 10 minutes followed by blocking in 1% (m/v) BSA in PBS. The

cells were then incubated in 4°C overnight with rabbit polyclonal antibody to α-tubulin

(1:1000, Abcam, Cambridge, MA) in 1% BSA containing solution, washed with PBS and

then incubated with Goat Anti-Rabbit IgG (H+L) antibody (1:500, Life Technologies) at

room temperature for 1 hour.

Protein Silencing

Short-hairpin RNAs targeted to SUN2 (5’-tcggatcttcctcaggctatt-3’), Nesprin-2G

3’UTR (5’-gcacgtaaatgacctatat-3’) or Luciferase (5’-gtgcgttgttagtactaatcctattt-3’) were

PCR and cloned into retroviral plasmid vector. 293T cells were transfected with the

plasmid and pseudo-typed envelope proteins to make virus by calcium phosphate

transfection. Viruses were harvested 24 hrs after transfection. Then NIH 3T3 cells

(0.5x105 cells/12 well) were infected by virus (0.6 ml virus) with 4 µg/ml polybrene and

re-plated the second day. Cell culture media were changed to DBS DMEM overnight

before cell spreading assay

Western Blotting

Transfected samples were harvested with 1X Laemmli sample buffer and boiled

at 95 ˚C for 10 min. Amersham Protran Premium 0.2 NC (10600004 GE Healthcare,

Little Chalfont, United Kingdom) membrane was used for protein transference. Primary

antibodies to probe proteins of interest in Western Blot were rabbit anti-SUN2, rabbit

anti-Nesprin-2G [26], mouse anti-GAPDH (clone 6C5, Life Technologies, Carlsbad, CA),

Page 25: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

25

LI-COR IRDye 680RD donkey anti-rabbit IgG (926-68073) and IRDye 800CW donkey

anti-mouse IgG (926-32212) were used. Signals were detected by Odyssey LI-COR

system (LI-COR Biosciences, Lincoln, NE). Images were processed by ImageJ (NIH).

Microscopy and Image Analysis

Fixed cells were imaged on a Nikon A1 laser scanning confocal microscope

system (Nikon, Melville, NY) with 60X/1.40NA oil immersion objective. The live cell

imaging was conducted on the same system within the environment of 37°C and 5%

CO2. For measuring the nuclear height, z-stacks were taken at an interval of 0.3 µm

and the x-z view projections were reconstructed using the NIS Elements application

(Nikon, Melville, NY). The maximum projection intensity analysis was applied to the x-z

images of the stained nucleus, and the top and bottom edges of the nucleus were

determined with the full width at half maximum (FWHM) technique [50] in MATLAB (The

MathWorks, Natick, MA). The height was calculated as the distance between the top

and bottom nuclear edge. Nuclear x-y dimensions (major and minor axis) were

measured using ImageJ (NIH). The aspect ratio was calculated as the height divided by

the length of the major axis in the x-y plan. The nuclear volume measurements were

performed using Volocity Demo (Perkin Elmer, Akron, Ohio).

Results

Collapse of Apical Nuclear Surface Contributes to Nuclear Flattening during Early Cell Spreading

We used x-z laser scanning confocal fluorescence microscopy of NIH 3T3

fibroblasts expressing GFP-histone to prepare time-lapse images of the nucleus as they

settled from suspension onto a fibronectin-coated glass dish. Three distinct nuclear

behaviors could in general be discerned during the spreading. First, the nucleus

Page 26: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

26

translated toward the base of the cell and the lower surface of the nucleus began to

flatten against the substratum in the first few minutes of attachment (Figure 1-1A). The

speed of initial translation of the nucleus toward the substratum was surprisingly fast --

about twenty-fold faster than would be expected from gravitational settling (based on

the assumed cytoskeleton viscosity in Table A1). Next, the top surface of the nucleus

collapsed while the length of the flattened bottom surface stayed roughly constant. This

change in the nuclear shape happened over a duration of 5-6 minutes (Figure 1-1B). In

the third phase, the collapsed nucleus increased in width at nearly constant height

(Figure 1-1C; see Figure 1-1D for quantification of the shape in Figure 1-1A, B and C;

these findings were consistent among five other live cell imaging experiments; see

Figure 2-2 A and B for examples). By constructing x-z nuclear shapes from different

view angles, we confirmed that the flat nuclear shapes were due to deformation of the

nucleus instead of already-flat nuclei toppling onto their sides (Figure 2-2). In addition,

by inverting the substrate on which the cells spread, we confirmed that gravity did not

affect the initial nuclear translation to the substratum nor the rate of the subsequent

flattening (Figure 2-3 B-D). In addition, nuclear flattening only occurred during cell

spreading; inhibition of actin assembly and cell spreading by cytochalasin D or

latrunculin A prevented nuclear flattening, as quantified by the aspect ratio (height/major

axis length) (Figure 2-4 A-C).

Although cells spread continuously in the first 60 minutes increasing their spread

area from less than 200 μm2 to nearly 1400 μm2 (Figure 2-5A and B), the nuclei

flattened and reached a steady-state height early during cell spreading in the first 20-30

minutes, when the cell itself had only spread to less than 50% of its final area (Figure 2-

Page 27: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

27

5A and B). Nuclear width stayed roughly constant over the first 20 minutes (Figure 2-

5C) and then increased steadily as the cell spread. As seen in Figure 2-5D, the aspect

ratio decreased to around 0.25 by 30 minutes, indicative of a ‘flat’ nucleus with a width

that is four times its height. We also observed that there was a separation between the

nuclear surface and the cell membrane (Figure 2-6A and B), which increased slightly

and then decreased over time as the cell spread and the nucleus flattened (reaching a

peak separation of around 2μm). The increase in the separation coincided with the

collapse of the top nuclear surface (compare Figure 2-5B and 2-6B).

Nuclear Flattening does Not Require Actomyosin Contraction in Spreading Cells

We pre-treated well-spread cells with three different inhibitors: Y-27632, a ROCK

inhibitor, ML-7, a myosin light chain kinase (MLCK) inhibitor or blebbistatin, a direct

inhibitor of myosin II activity. Our approach was to pretreat cells for 1 hour at the

appropriate dose, trypsinize cells, and to allow them to attach for 1 hour in medium

containing the inhibitor (Figure 2-7A). Neither blebbistatin (50 µM) nor Y-27632 (25 µM)

interfered with the nuclear flattening process observed in normal cells (Figure 2-7A-B,

E-H), suggesting that myosin II activity is not required for nuclear flattening. However,

ML-7 (25 µM) treatment interfered with both nuclear flattening and cell spreading

(Figure 2-7A and B). To understand the differential effects of the drugs on nuclear

flattening, we measured the area of cell spreading (Figure 2-7C) and correlated nuclear

aspect ratio with the cell spreading area (Figure 2-7D; correlations between nuclear

height and width with cell spreading area are in Figure 2-7G and H). The degree of cell

spreading decreased dramatically in ML-7 cells (Figure 2-7C and D). This revealed a

potential reason for the differential effects: ML-7 treatment prevented cell spreading

while Y-27632 or blebbistatin treatment altered cell shapes but did not prevent cell

Page 28: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

28

spreading. Nuclear aspect ratios and cell spreading areas were comparable in

untreated control cells at 15 minutes (Figure 2-5D) and ML-7 treated cells (Figure 2-7B

and C). Taken together, these data suggest that the degree of cell spreading appears to

be a predictor of nuclear flattening in myosin inhibited cells. We and others have shown

in the past that inhibiting myosin activity in well-spread cells rounds the nucleus [23, 24].

In consideration of the above results, we examined the effect of the three myosin

inhibitors on nuclear height in well-spread cells. Myosin inhibition again resulted in a

rounded nucleus only when the cell was rounded by the action of the drug (Figure 2-

8A). In these experiments, blebbistatin (but not Y-27632 and ML-7) treatment resulted in

rounded cell morphologies; only blebbistatin treated cells showed rounded nuclear x-z

cross-sections (Figure 2-8 B-H, correlations between nuclear height and width with cell

spreading area are in Figure 2-8 G and H).

We next examined whether myosin inhibition altered aspects of nuclear

flattening, such as the initial collapse of the top surface. Inhibiting myosin with Y-27632

did not change the qualitative nature of the drop in nuclear height (except for an initial

lag time where the Y-27632 treated cell is unable to spread and the nucleus doesn’t

flatten in that time, Figure 2-9 A) The distance between the apical cell surface and

apical surface of the nucleus during collapse of the top surface increased significantly

more than control cells to a maximum of around 4μm (Figure 2-9 B). Hence, myosin

inhibition did not produce qualitative changes in the nuclear spreading dynamics. We

did find that inhibiting myosin with Y-27632 decreased the width of the flattened nucleus

and its volume (Figure 2-9 C and D) by a measureable amount. Thus, while the myosin

inhibition does not alter nuclear flattening, it appears that actomyosin forces may

Page 29: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

29

contribute to some increase in nuclear volume by widening the nucleus after the initial

flattening (Figure 2-9 D).

We note that the differential effects of ML-7 on initial cell spreading (which it

prevents) and on already spread cells can be explained by its differential effects on

retrograde flow in slow versus fast moving cells as shown by Jurado et al. [51]. In

spreading cells where adhesions are relatively smaller, ML-7 treatment causes

complete disassembly of adhesions and is predicted to increase retrograde flow and

prevent spreading, while in well-spread cells, the retrograde flow would be reduced due

to a decrease in the “raking” of adhesions. Blebbistatin treatment prevented cell

spreading and nuclear flattening if cells were allowed to spread for longer time (6 hours,

Figure 2-10 A-C); thus, blebbistatin uniformly appears to decrease retrograde flow but

presumably it inhibits adhesions at a slower rate, which would explain why it does not

completely inhibit initial cell spreading at one hour.

Intermediate Filaments and Microtubules are Dispensable for Nuclear Flattening in Spreading Cells

Given that actomyosin contraction was not required for flattening, but instead

nuclear flattening correlated strongly with the degree of cell spreading, we examined the

role of the other two cytoskeletal structures in the cell: intermediate filaments and

microtubules. Nuclear aspect ratio was measured and compared between vimentin +/+

mouse embryonic fibroblasts (vim +/+ MEFs) and vim -/- MEFs. Compared to control

cells, the nucleus was more rounded (although it was still significantly flattened to an

aspect ratio of 0.3) in vim -/- MEF cells after one hour of cell spreading (Figure 2-11A

and B), but consistent with our observations above, the cell was comparatively less

spread (Figure 2-11C). Importantly, over longer time (12hours), the nucleus was

Page 30: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

30

flattened in vim-/- cells (Figure 2-11B). Thus, while the absence of vimentin intermediate

filaments reduced the rate of nuclear flattening, it did not influence the extent of nuclear

flattening. To test if myosin activity was causing nuclear flattening in vim-/- MEFs, we

inhibited myosin activity in these cells with Y-27632. The nucleus was flattened in

myosin inhibited vim-/- MEFs (Figure 2-11B). Together, these results suggest that the

nucleus can flatten in the absence of myosin activity and intermediate filaments. The

experiments with these cells again point to a strong correlation between the spreading

area of the cell and nuclear flattening.

To determine the role of microtubules in nuclear flattening, we disrupted

microtubules with nocodazole and colcemid. At a dose of 1.65 μM nocodazole,

microtubules were eliminated from the cell, but the cell was unable to spread one hour

after seeding, and concomitantly, the nucleus did not flatten (Figure 2-12 A-C). Upon

decreasing the dose to 0.83μM, microtubules were greatly fragmented, but the cell was

not spread as well as the control cells at 1 hour. At 6 hours into the spreading process,

cells had no discernible microtubules (Figure 2-13A) but were able to spread (Figure 2-

13C) and the nuclei were flat (Figure 2-13B). We next allowed cells to spread overnight

and treated cells with 0.27μM colcemid. The treatment did not cause cell rounding

(Figure 2-13B) even though microtubules were completely disrupted (Figure 2-13A) and

the nucleus remained flat (Figure 2-13B). Together, both these results suggest that

microtubules are not required for nuclear flattening when cells are able to spread.

Apical and Basal Actomyosin Bundles are Not Required for Nuclear Flattening during Initial Cell Spreading

Actomyosin stress fibers have been implicated in shaping the nucleus [22].

Considering our results above which suggest that myosin activity is not required for

Page 31: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

31

nuclear flattening, we examined the presence of actomyosin bundles in spreading cells.

In the first 20 minutes when the nucleus flattened significantly, the average number of

actomyosin bundles above the nucleus was found to be approximately 0.2, i.e. 1 out 5

cells have one apical bundle (Figure 2-14A). The number of basal fibers under the

nucleus coinciding with the time of nuclear flattening was about 1 ~ 2 per cell (Figure 2-

14B). We next examined the correlation between the nuclear height and the number of

apical and basal bundles at different times in the cell spreading process. Figure 2-14C

shows three examples where neither basal nor apical fibers could be discerned during

initial cell spreading, although the nucleus had been flattened to a considerable extent.

As seen in the plots, there were several cells where no fibers are discernible (apical or

basal), but the nucleus is clearly flat (Figure 2-14 D and E). These results, combined

with the myosin inhibition experiments above argue against a mechanical explanation in

which apical or basal actomyosin bundles play a significant role in flattening the nucleus

during initial cell spreading.

Nuclear Flattening can be Reversed by Detachment of the Cell from the Substratum

The different experiments described above seem to consistently indicate that

nuclear flattening is strongly correlated with the extent of cell spreading. A rounded cell

is predicted to have a rounded nuclear x-z cross-section, while a well-spread cell is

expected to have flat nucleus. To further test the relationship between the degree of cell

spreading and nuclear height, we treated well-spread cells with trypsin and measured

the nuclear x-z cross-section. At concentrations of trypsin (0.25% w/v) normally used for

cell passage, the nucleus rounded up remarkably fast (in a few seconds) coupled with

fast cell rounding (Figure 2-15A). There was a strong relationship between the degree

Page 32: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

32

of cell rounding as measured by the contact length between the cell and the surface of

the substratum, and the nuclear height, at different times during the trypsinization

process (Figure 2-15B). We next treated cells with trypsin at reduced concentrations

(0.08% w/v). This slowed the cell rounding process significantly (several minutes).

Consistent with the expectation that nuclear height is determined primarily by the

degree to which the cell is spread, the nucleus did not round until the cell had

significantly changed its shape through release of cell-substratum adhesions (Figure 2-

15C). This occurred over several minutes (Figure 2-15D). These results strongly

support the concept that nuclear height correlates with the degree of cell spreading.

The LINC Complex is Not Required for Nuclear Flattening

The LINC (for Linker of Nucleoskeleton to the Cytoskeleton) complex has been

shown to transmit mechanical forces from the cytoskeleton to the nucleus [34]. We

therefore asked if an intact LINC complex is required for nuclear flattening. The

disruption of the LINC complex by over-expression of GFP-KASH4 (KASH4 is the

KASH domain from nesprin 4 which competitively binds to the SUN proteins, but lacks

the cytoskeletal linker domain) [27] slowed the flattening of the nucleus (Figure 2-16A

and B) but it also slowed normal spreading of the cells (see also Figure 2-16G and H).

Importantly, at 6 hours and 24 hours (Figure 2-17 A-C), GFP-KASH4 expressing cells

were well-spread and displayed flat nuclei. Similarly, the knockdown (Figure 2-17 D) of

nesprin 2G (Figure 2-16C and D) and SUN2 (Figure 2-16E and F) with shRNA

interference did not have any effect on nuclear flattening during initial cell spreading

(see Figure 2-16G and H for a statistical comparison of all the data). The data indicates

that an intact LINC complex is not required for nuclear flattening and cell spreading.

Page 33: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

33

We next examined the effect of lamin A/C on the degree of nuclear flattening

(Figure 2-18). At 60 minutes, lamin A/C -/- MEFs had more flattened nuclei compared

to WT MEFs; however, WT MEFs did not spread significantly at 60 minutes (Figure 2-

18A). When allowed 6 hours to spread however, WT MEFs were able to spread and

flatten their nuclei. These results suggest that the absence of lamin A/C correlates with

an increased rate of nuclear flattening, leading to a flattened nucleus in lamin A/C -/-

MEFs compared to WT MEFs during cell spreading.

A Mathematical Model for Nuclear Flattening and Cell Spreading

The presence of individual cytoskeletal elements (microtubules, intermediate

filaments), myosin activity, or an intact LINC complex, which transmits forces from the

cytoskeleton to the nucleus, is not required for flattening the nucleus as long as the cell

is able to spread. We found that inhibiting F-actin polymerization which prevents cell

spreading prevented nuclear flattening (Figure 2-4A-C). Thus, nuclear flattening

correlates with the degree of cell spreading. Based on these results, we propose a

simple mechanical model which shows that stresses arising from cellular shape

changes and cytoskeletal network assembly from the apical cell cortex are sufficient to

explain nuclear translation to the surface and flattening against the substratum. We

modeled the cell’s cytomatrix, i.e. the cytoskeletal network phase connecting the

nucleus to the cell membrane, as a contractile compressible material that resists

compression/expansion and shear strains (like the approach by Dembo and coworkers

[52-54]). On the slow time scale of spreading (several minutes), only the viscous

resistance to deformation is considered relevant (i.e. elastic forces are considered

negligible given the remodeling that occurs in the cell over long time scales), such that

the stress tensor is proportional to the rate-of-strain tensor, i.e.

Page 34: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

34

𝛔 = 2𝜇�̇� + σ𝐜𝐈 (2-1)

Here σc is the contractile stress due to myosin motor activity, 𝐈 is the unit dyadic,

ε̇ = 1

2(∇v + ∇vT) is the rate-of-strain tensor, and μ is viscosity which measures the

modulation of stress due to both expansion/compression and shear deformations of the

compressible network phase. Note that both shear and expansion/compression modes

in ε̇ are relevant since the network is assumed compressible. Equation (2-1) can be

considered a slow-flow limit of the more general two-phase reactive interpenetrating

flow models for cells developed by Dembo and coworkers ([52-54]) where network

contractile/viscous properties can be assumed to be uniform and hydrostatic pressure

gradients are assumed negligible.

Solving the momentum balance ∇ ∙ σ = 0 with the appropriate boundary

conditions yields the stress and velocity fields of the network. Before we discuss the full

general model for cell and nuclear mechanics during nuclear flattening, we show a

simple model which illustrates the key predictions of the general model. The simple

model (Figure 2-18A) is an approximate representation of the gap between the cell apex

and the nuclear apical surface when the gap is small compared to the inverse curvature

of the nucleus. The main purpose of this model is to show how movements of the top

cell membrane and flow from the membrane of network can exert a stress on the

nuclear surface. To illustrate the relevant properties of a cell containing

contractile/viscous medium obeying Equation (2-1), consider a simplified one-

dimensional case illustrated in Figure 2-18A, which represents the planar approximation

of the local gap of length L between the cell membrane and the nuclear envelope (the

exact derivation for a spherical cell is presented in in the Materials and Methods). Let

Page 35: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

35

the gap expand at speed V by moving the cell membrane and keeping the nuclear

surface fixed, and assume new network assembles at the cell membrane (where f-actin

is primarily generated) at speed va. In the special case where V = va, there is no

network flow because the network assembles at exactly the rate required to fill the

volume behind the moving membrane. Otherwise, there will be network expansion V >

va or compression V < va, either of which will modulate the stress on the nuclear surface

at the base. As derived in the Materials and Methods, the resulting velocity and stress

fields for the 1-D approximation are:

vx = (V − va)x

L, (2-2)

σxx = σc + 2μdv

dx= σc + 2(V − va)

μ

L. (2-3)

Since stress is uniform in this case, the tensile stress on the nuclear surface is

equal to σxx. From equation (2-2) and (2-3), the following important properties regarding

transmission of stress to the nuclear surface are evident: (1) expansion of the gap V > 0

between the cell membrane and the nuclear envelope will increase the net tensile stress

σxx on the nuclear surface; and (2) compression of the gap (V<0) or assembly of

network at the cell membrane (va > 0) will decrease the net tension on the nuclear

surface. An important corollary to these predictions is that when the nuclear surface

stress is fixed (instead of fixing the nuclear surface position), the nuclear surface will

move at a speed such that the gap expansion speed V satisfies the stress balance in

Eq.(2-3). For example, if the nuclear surface stress is in balance with the network

contractile tension, such that σxx = σc, then V = va. Consequently, the nuclear surface

will move together with the membrane keeping V = 0 when va = 0, or it will move away

from the cell membrane at speed va when va > 0. In this way, the nuclear surface

Page 36: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

36

movements will tend to follow the movements of the nearby cell membrane boundary,

but will also tend to move away from cell membrane surfaces where network is being

assembled. These are the important properties of the network that govern the more

general model that now follows for nuclear shape changes during cell spreading.

To model the case of a spreading cell (Figure 2-19B), new network is assumed to

assemble where F-actin is generated at the cortex and at the cell edge on the

substratum, but not at any other substratum-cell membrane interface (See Materials

and Methods for model and simulation details). Throughout the network phase, F-actin

and the other constituents of the cytoskeletal network (intermediate filaments,

microtubules) are assumed to assemble/dissemble to re-equilibrate the density and

mechanical properties of the network relatively quickly on the slow time scale of cell

spreading. As shown in Figure 2-19C, these assumptions and the constitutive stress

equation (Eq.2-1) are sufficient to predict cell spreading very similar to the experimental

observations, including the observed initial distension and net translation of the nucleus

toward the substratum and initial flattening against the substratum. Cell spreading is the

result of assembly of network at the contact boundary, which generates a centripetal

flow of network. Substratum adhesion hinders this centripetal flow, resulting in a net

outward expansion of the cell boundary near the substratum. Expansion near the

substratum corresponds to retraction of the upper cell surface away from the

substratum in order to preserve cell volume (assumed constant due to the cell’s osmotic

resistance to volume changes). Because of viscous resistance to network expansion

(Eq. 2-1), the movement of the cell boundaries generates stress on the nucleus as the

intervening network expands or compresses, and movements of the nucleus boundary

Page 37: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

37

tend to follow those of the cell boundaries. Assembly of new network at the cortex has

the effect of increasing the downward compressive flow of network. Because the

network is assumed not to assemble at the cell-substratum interface (except at the

contact boundary), this flow causes an initial vertical distension of the nucleus followed

by a net translation of the nucleus toward the substratum (Figure 2-1C). While network

assembly at the cortex is required to predict the initial rapid downward translation of the

nucleus given the assumed network viscosity, cell spreading and the resulting nuclear

flattening only requires an assumption of network assembly at the contact boundary, as

discussed below. As shown in Figure 2-19D, the time-dependent nucleus height and

width predicted by this model agree well with the experimentally observed trends.

As detailed in the Materials and Methods, our mechanical model of the nucleus

accounts for resistance to compression, and a resistance to nuclear envelope

expansion which accounts for an excess of surface area of the nuclear lamin network

above that of a smooth sphere with the same volume. This excess surface area is

evident from the observed undulations in the lamin network [55]. As shown in Figure 2-

19E, the nuclear volume remained nearly constant during cell spreading, but the

apparent surface area of the nuclear envelope increased; once the surface area

expansion approached the “true” surface area, further shape changes were minimal due

to the large mechanical resistance to further surface area changes. Hence, a steady-

state nuclear shape is reached before spreading stops, consistent with our

observations, and the steady-state shape of the flattened nucleus depends primarily on

the stiffness of the nuclear lamina and the excess surface area of the initially rounded

nucleus. Without the assumption of network flow from the cortex, the nucleus is still

Page 38: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

38

predicted to flatten as the cell spreads due to the vertical compression and horizontal

expansion arising from the moving cell boundaries (Figure 2-20A). However,

reproducing the relatively rapid approach and flattening of the nucleus against the

substratum early in cell spreading processes requires an assumption of network

assembly and flow from the apical cell cortex. Although the initial nuclear dynamics are

similar, the fully spread model cell with or without apical cortical network assembly

appear very similar at longer times. Hence, our results suggest that apical cortical

network assembly and flow is necessary to translate the nucleus to the substratum early

in spreading, but it is not required to explain the ultimate flat nuclear shapes like those

observed in experiments. Reproducing the observed flattening dynamics therefore

does not require the assumption of continued network assembly at the apical cortex at

longer time (< ~25 min).

When the substratum adhesion frictional parameter is reduced, the speed of

retrograde flow near the substratum increases (consistent with the molecular clutch

model [56]). At a sufficiently low adhesion, cell spreading slows and stops at a steady

state before the cell can fully spread, and the nucleus also stops flattening when the cell

stops spreading (Figure 2-20B). This result reinforces the prediction that nucleus shape

changes tend to follow cell shape changes.

Interestingly, the predicted dynamics of nucleus spreading do not depend

significantly on the background tension of network, as shown in the simulation results in

Figure 2-20C, where the network tension parameter σc was set to zero. The differential

stresses that cause nuclear shape changes arise primarily from the resistance to

expansion or compression of the network, not from the background contractile tension

Page 39: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

39

of the network, which is treated here as a uniform tension that acts equally on all

surfaces. The predicted lack of dependence on contractile tension is consistent with our

experimental observation that inhibition of myosin does not prevent nucleus flattening in

spreading cells. (It should be noted, however, that if σc were to vary spatially, the

contractility gradient (∇σc) would drive local network flow in the gradient direction.)

As mentioned above, the shape of the flattened nucleus depends on the area

stiffness of the nuclear surface. When the area modulus was set to zero, the nucleus

continued to flatten as long as the cell continued spreading (Figure 2-20D). This

predicted behavior is consistent with the increased nuclear flattening in lamin A/C -/-

MEFs (Figure 2-18). Since the nucleus can flatten without changing volume, the

dynamics of nucleus flattening did not significantly depend on the value of volume (bulk)

modulus of the nucleus (see appendix for a fuller discussion of sensitivity of the

predictions to model parameters).

In summary, the key predictions of the model and simulations are (1)

distension/translation of the nucleus toward the surface is driven by assembly of actin at

the apical cortex, (2) nuclear flattening is driven by stresses caused by cytoskeletal

network expansion/compression upon movement of the cell boundaries, and (3) these

nuclear shape changes can arise without network contractile tension or stress fibers.

The model predictions therefore provide an explanation for the experimental

observations of nuclear flattening against the substratum without significant actomyosin

contractile tension.

Discussion

The flattened nucleus is a common feature of cultured cells, but the mechanisms

by which it is flattened have remained obscure. There is mounting evidence that the

Page 40: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

40

cytoskeleton exerts forces on the nucleus to position it [29, 57-59]. In this paper

however, we show that as long as the cell was able to spread, inhibiting actomyosin

forces, microtubule-based forces and intermediate filaments, as well as the LINC

complex, did not prevent nuclear flattening. Remarkably, nuclear height correlated

tightly with the degree of cell spreading. Independent of the type of cytoskeletal force

perturbed, the nucleus is flat unless the perturbation prevents initial cell spreading, or

rounds a spread cell.

This robust feature of nuclear shaping suggests that it is the dynamic

deformation of the cell shape itself that causes nuclear flattening consistent with our

previous results reporting reversible nuclear deformation caused by proximal cell

protrusions in migrating cells [37]. The fact that the nuclear apex collapses during the

nuclear flattening, opening up a significant distance between the cell apex and the

nuclear apex (on the order of a few microns), argues against the cell cortex directly

compressing the nucleus downward. The near complete absence of apical actomyosin

bundles argues against any explanation for flattening that requires a downward

compressive force on the nuclear apex by large actomyosin bundles (for example ref.

[60]). That apical fibers do not participate in the flattening process does not argue

against later distortion of the nucleus by fully developed actomyosin bundles as

reported by others [61].

Neither intermediate filaments nor microtubules are required for flattening.

Finally, disruption of the LINC complex via KASH4 over-expression failed to prevent

nuclear flattening. It slowed the rate of cell spreading, suggesting perhaps that a

coupled nuclear-cytoskeleton is required for rapid F-actin polymerization, but it did not

Page 41: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

41

prevent flattening over longer times. Given that myosin activity, MTs and vimentin IFs

are not required, that the LINC complex is dispensable is perhaps not surprising. We

have shown before that KASH4 over-expression results in rounded nuclear shapes in

cells on polyacrylamide gels [62]. This difference may be due to possibly different cell

spreading dynamics on gels versus glass. We note that the cell spreading area in

KASH4 cells was lower on gels, suggesting that the relationship between nuclear

flattening and cell spreading is conserved on other types of surfaces.

Our computational model demonstrates that expansive/compressive stresses

arising from movement of the cell boundaries and centripetal flow of cytoskeletal

network from the cell membrane is sufficient to explain translation of the nucleus toward

the substratum and subsequent flattening against the substratum. The fact that the

experimentally observed flattening dynamics could be closely reproduced using one

constitutive equation (Eq.2-1), a simple model for cell mechanics, and one “fitted”

parameter 𝑣𝑎 provides strong support for the validity of the model assumptions.

Moreover, the model successfully predicts several experimental findings: the approach

to a steady-state flattened nuclear shape despite continued cell spreading, nuclear

flattening in the absence of actomyosin tension, increased nuclear flattening in the

absence of lamin A/C, and the cessation of nuclear flattening upon the cessation of cell

spreading.

Consistent with the assumption that the nucleus is under tension, we found that

the volume of the nucleus decreased in myosin-inhibited cells. However, our

experiments show that flattening is not a consequence of tension. As explained by the

computational model, flattening can instead arise from the motion of the cell boundary

Page 42: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

42

transmitting stresses to the nuclear surface because the intervening cytoskeletal

network resists expansion or compression. As a result, the nuclear shape changes tend

to mimic changes in cell shape during cell spreading.

Interestingly, the presence of actomyosin contraction in normal cells does not

alter the dynamics of nuclear shape changes during cell spreading. In the presence of

contraction, the net stress on the nuclear surface in the absence of any F-actin

assembly from the membrane (such as in serum-starved cells) is likely tensile.

However, even if this stress in the network is net compressive (such as when myosin is

inhibited), the differential stresses between apex and sides of the nucleus that drive

nuclear shape dynamics during cell spreading are predicted to be similar (Figure 10C).

In summary, our results support a surprisingly simple mechanical system in cells

for establishing nuclear shapes. Our computational model suggests that nuclear shape

changes result from transmission of stress from the moving cell boundary to the nuclear

surface due to frictional resistance to expansion/compression of the intervening

cytoskeletal network. Nuclear shaping are thus driven by cell shape changes.

Page 43: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

43

Figure 2-1. The dynamics of nuclear flattening during early cell spreading. A-C). Shown are vertical cross-sections of a nucleus in a cell that settles and spreads on the substratum. The images were captured using x-z laser scanning confocal fluorescence microscopy; the nucleus is expressing GFP-histone H1. Three phases were discernible in the nuclear flattening process: A). A settling phase where the basal surface of the nucleus contacted the basal cell surface and started to spread, the height was roughly constant during this time. B). A collapse of the top surface where the basal surface of the nucleus did not spread much, and C). A widening phase where the basal surface of the nucleus continued to spread and contributed to nuclear widening; the height was roughly constant in this phase. Scale bar is 10 µm. D. Plot shows nuclear height and contact length in A-C with time.

Page 44: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

44

Figure 2-2. The nuclear deformation causes nuclear flattening but not already-flat

nucleus toppling onto their side. Images show x-z view of the nuclear shape from two different view angles in two cells at different time. Scale bar is 5 µm.

Page 45: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

45

Figure 2-3. The dynamics of nuclear flattening against substratum is not influenced by

gravity. A) Nuclear shape changes during cell spreading are shown for normal (top) and inverted samples (bottom) at different time points in x-z view. For the inverted case, cells were allowed to first settle and attach for 5min before inverting the sample. Neither nuclear aspect ratio B) nor cell spreading area C) has a significant difference between control and inverted

(n ≥ 32). Scale bar is 10 µm. All data are shown as Mean ± SEM

Page 46: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

46

Figure 2-4. Nuclei does not flatten when cell spreading is prevented by inhibitors of

actin assembly. A) Images show the x-y and x-z view of control cell and cell treated with cytochalasin-D or latrunculin-A. B) Nuclear aspect ratio (height divided by the length of the major-axis in the x-y plane) increases significantly after 1hr spreading in the presence of the drugs, which corresponds to the decreases of cell spreading area C). (n ≥ 31, * indicates p<0.05; all comparisons are with untreated control). Scale bar in (A) is 20 µm in the x-y view and 5 µm in x-z view. All data are shown as Mean ± SEM.

Page 47: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

47

Figure 2-5. The nucleus flattens completely in a partially spread cell. A) Images show

cells at different stages in the spreading process. Nuclei (blue) were completely flattened at roughly 20-30 minutes when the cells (green F-actin) had not spread completely. Scale bar is 20 µm for the x-y views, and 10 µm for the x-z views. Also shown are average nuclear heights B), average nuclear widths C) and nuclear aspect ratio (height/width) D) at different times during the cell spreading process, and the corresponding areas of cell spreading. The nuclear heights reach an approximate steady state at around 30 minutes, when the cells spread to about 50% of the final

area (n ≥ 32 cells).

Page 48: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

48

Figure 2-6. The apical surface of cell was separated from the apical surface of nucleus in the early stage of cell spreading. A) Images of nucleus (blue) and cell (green) show the gap between them in the vertical cross-sections at different time of cell spreading. Scale bar is 10 µm. B) The gap between the top cell

surface and the top nuclear surface plotted with time (n ≥ 25 cells). The gap

increased at the beginning reflecting the collapse of the nuclear surface and

then decreased over time to near zero levels. All data are shown as Mean ±

SEM.

Page 49: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

49

Figure 2-7. Nuclear flattening is independent of actomyosin contraction. A) Images show cells pre-treated with drug for 1 hour, trypsinized, and then seeded onto substrates for 1 hour in the presence of the drug. Cells in the presence of Y-27632 and blebbistatin showed clear effects on the cell morphology compared to the control, but the nucleus was still flattened as evident from the x-z cross-section. ML-7 treatment on the other hand prevented the spreading of the cell as well as the flattening of the nucleus. Scale bar for x-y views is 20 µm, for x-z views is 5 µm. B) Nuclear aspect ratio was larger in ML-7 treated cells reflecting unflattened nuclei, consistent with the fact that the cells were unable to spread in presence of ML-7 C). Minor differences in aspect ratio on Y-27632 or blebbistatin treatment reflect minor effects on degree of cell spreading. D) Aspect ratio correlates with the degree of cell spreading. In ML-7 treated cells, the cells were unable to spread (blue diamonds) corresponding to the large aspect ratio; none of the other treatments prevented nuclear flattening but concomitantly, they did not prevent cell spreading. Bar plots shown are nuclear height E) and width(F) during initial cell spreading under myosin inhibition and the corresponding

scatter plot G) and (F), (n ≥ 31 for all conditions, * indicates p<0.05; all

comparisons are with untreated controls).

Page 50: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

50

Page 51: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

51

Figure 2-8. The effect of myosin inhibition on nuclear shape in well spread cell. The experiments in A-D) show the results on treating well-spread cells (cultured overnight on fibronectin coated glass-bottom dishes) with myosin inhibitors. Blebbistatin treatment rounded up spread cells, and caused rounded nuclear shapes. Images are shown in A), while the scatter plots of aspect ratio versus cell spreading area are shown in D). B) and C) show the average aspect ratio and areas for the different myosin inhibitors. Shown are nuclear height E) and width F) of well-spread cell with myosin inhibition, and their corresponding scatter plot with cell spreading area G) and H). Scale bar in A is 20 µm for the

x-y view and 5 µm for the x-z view. (n ≥ 24, * indicates p<0.05; all

comparisons are with untreated control. All data are shown as Mean ± SEM)

Page 52: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

52

Page 53: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

53

Figure 2-9. Inhibition of myosin activity with Y-27632 does not alter the qualitative

features of dynamic nuclear flattening during cell spreading. As seen in A),

the time-dependent changes in nuclear height (n ≥ 33) still occur on Y-

27632 pre-treatment while the cells spread (an initial lag time in the flattening of the nucleus is attributable to an initial lag time in the spreading). Y-27632 treatment increased the separation between the apical

cell and nuclear surfaces B) (n ≥ 29). The decrease in nuclear width C)

and volume D) in Y-27632 treated cells suggests that myosin contraction

plays a role in the nuclear widening process (n ≥ 29). ( * indicates p<0.05;

all comparisons are with untreated control. Scale bar is 10 µm in both x-y view and 5 µm for the x-z view. All data are shown as Mean ± SEM)

Page 54: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

54

Figure 2-10. Blebbistatin prevents cell spreading at later times resulting in final

nuclear rounding. A) At 1 hr images show the spreading of blebbistatin-

treated cells (n ≥30) in x-y view and the associated nuclear height in x-z

view. Given more time (6 hours), the area of cell spreading decreased significantly C) and the nucleus rounded up to the similar level of well spread cell treat with blebbistatin. Consistent with this, nuclear aspect ratio at 6 hours was higher than control B). ( * indicates p<0.05; all comparisons are with untreated control. Scale bar is 10 µm in both x-y view and 5 µm for the x-z view. All data are shown as Mean ± SEM.)

Page 55: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

55

Figure 2-11. The absence of intermediate filaments does not prevent nuclear flattening during cell spreading. A) Intermediate filaments are not required for nuclear flattening during cell spreading. The nucleus is flattened in vim-/- cells similar to vim+/+ cells at 12 hours after cell seeding although vim-/- nuclei are slightly rounded at 1 hour into the spreading process. Y-27632 treatment in vim-/- cells did not prevent nuclear flattening. Scale bar in x-y view is 20 µm, in the x-z view is 5 µm. B) and C) show the average aspect ratio and spreading areas; all the differences in aspect ratio can be attributed to the corresponding (inversely related) differences in the degree

of cell spreading.( * indicates p<0.05, n ≥ 30)

Page 56: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

56

Figure 2-12. Disruption of microtubules by nocodazole rounds up the nucleus but also prevents cell spreading. (A) Immunofluorescence images are shown of 1.65 µM nocodazole treated cells in two kinds of experiments, one in which the drug was treated during initial cell spreading, and the other in which cells that were well-spread were treated with nocodazole. For both experiments, no distinct microtubules were visible, but nuclei were not flat as the cells had rounded morphologies. The quantifications of nuclear aspect ratio (B) and cell spreading area (C) again show that the nuclear

rounding is correlated with the extent of cell spreading.(n ≥ 30, * indicates

p<0.05; all comparisons are with untreated control. Scale bar is 10 µm in both x-y and x-z view. All data are shown as Mean ± SEM)

Page 57: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

57

Figure 2-13. The absence of microtubule does not prevent nuclear flattening during cell

spreading. A) Shown is the effect of nocodazole (0.83 μM) on nuclear flattening and cell spreading, and the effect of colcemid (0.27μM) on well spread cells. At 6 hours, the nocodazole-treated cells were spread and had flattened nuclei, while no microtubules were visible. Likewise, colcemid treatment for 1 hour disrupted microtubules in originally well-spread cells but did not alter nuclear height. Collectively the data suggests that microtubules are not required to establish or maintain a flattened nucleus. Measurements of aspect ratio B) and spreading area of the cells C) under various conditions.

n ≥ 30, * indicates p<0.05; all comparisons are with untreated control. Scale

bar in (E) is 10 µm for x-y view and 5 µm for x-z view. All data are shown as Mean ± SEM.

Page 58: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

58

Figure 2-14. Apical and basal actomyosin bundles are not required for nuclear flattening during initial cell spreading. A) Apical actomyosin bundles were counted above the nucleus in spreading cells (actin cables were visualized by phalloidin staining). Apical bundles appear only after 30-40 minutes by which

time the nucleus has flattened completely (n≥30). B) In spreading cells, one

basal actomyosin bundle on average appeared under the nucleus by around 15 minutes. Basal cables were only counted if they ran beneath the nucleus.

Times represent time after initial seeding, (n≥30). C) Images show examples

at different times after seeding of apical and basal F-actin stained cells (green) that lack actomyosin bundles, but have significantly flattened nuclei (blue). Scale bar is 10 µm for both panels. D) and E) show plots of the nuclear height with the number of apical and basal actomyosin bundles at 15, 20 and 30 minutes. A number of examples can be seen where there are zero apical

or basal actomyosin bundles but the nucleus is still significantly flattened (n≥

30 cells). All data are shown as Mean ± SEM.

Page 59: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

59

Courtesy David Lovett

Page 60: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

60

Figure 2-15. Nuclear flattening can be reversed by detachment of the cell from the

substratum. Trypsinization of cells rounded the nucleus A) in a remarkably short time of a few seconds. Importantly, the nuclear rounding closely followed the cell rounding- the dynamics of height changes (gray circles) and changes in contact length of the basal cell surface (black circles) are similar B). Scale bar is 10 µm for both panels in A. This concept was tested further in C) by trypsinizing cells at 1/3 the dose of the trypsin concentration used in A. The nucleus rounded much more slowly (several minutes) and closely reflected the rounding up of the cell body (the nuclear height and cell contact length are shown in D)). Thus, the degree of cell spreading determines the degree of nuclear rounding during cell detachment. Scale bar is 20 µm for both panels. All data are shown as Mean ± SEM.

Page 61: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

61

Figure 2-16. The LINC complex is not required for nuclear flattening during cell spreading. A) GFP-KASH4 expression in cells prevented flattening of nuclei at 1 hour, but also prevented cell spreading. 6 hours into the spreading, the nucleus did flatten in GFP-KASH4 expressing cells. Scale bar in x-y and x-z views are both 20 µm. B) shows the scatter plots of nuclear aspect ratio versus cell spreading area in GFP expressing (control) versus GFP-KASH4 expressing cells; GFP-KASH4 expressing cells did not spread well at 1 hour after seeding, which correlated with the expected response of a lack of nuclear flattening. C), D) Nesprin 2 knockdown did not produce any effects on aspect ratio nor the degree of cell spreading. E), F) SUN2 knockdown produced no effects on aspect ratio and degree of cell spreading. Scale bar in x-y view is 20 µm and x-z view is 5 µm in C and E. G and H show comparisons of average aspect ratio, and cell spreading area at the different conditions. Only when the cell is not able to spread does the nucleus remain

unflattened (* indicates p<0.05, n ≥ 35). All data are shown as Mean ±

SEM.

Page 62: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

62

Page 63: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

63

Figure 2-17. GFP-KASH4 overexpression slows down but does not prevent cell spreading and nuclear flattening. A) Images show GFP-KASH4 cells after one hour of spreading have unflattened nuclei and not so-well spread cells; but nuclei become flat at long times (6 and 24 hours) with fully spread cells. The changes of nuclear aspect ratio and cell spreading area in B) and C) respectively, indicate that the rounded nucleus in cells expressing GFP-KASH4 at 1 hour is correlated with a slower cell spreading rate. D). Western blots of indicated proteins immunoprecipitated by SUN2 or Nesprin 2G

antibodies from NIH 3T3 cell lysates. (n ≥ 37, * indicates p<0.05). Scale bar

is 20 µm in both x-y and x-z view. All data are shown as Mean ± SEM.

Page 64: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

64

Page 65: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

65

Figure 2-18. Nuclei in lamin A/C -/- MEFs flatten faster than WT cells. Nuclei in WT MEFs are less flattened at 1 hour after cell seeding A) compared to lamin A/C -/- cells which have extremely flat nuclei. The degree of cell seeding is small in WT at 1 h and increases by 6 h; lamin A/C -/- cells however are well-spread at 1 h. Scale bar in the x-y view is 20 µm, in the x-z view it is 10 µm. The lack of cell spreading, higher nuclear heights and lower nuclear widths at 1 h in WT cells compared to lamin A/C -/- cells is evident in the scatter plots of nuclear aspect ratio versus cell spreading area B), as well in the average values of nuclear aspect ratio and spreading area (C and D). (* indicates

p<0.05; n ≥ 43; all comparisons are with WT cells at 1 hour spreading). All

data are shown as Mean ± SEM

Page 66: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

66

Page 67: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

67

Figure 2-19. Mathematical model for nuclear deformation during cell spreading. A) Predictions of a simplified one-dimensional model for the cytoskeletal network spanning the gap between the nuclear surface and the cell membrane. Movement of the membrane relative to the nuclear surface (speed V) or assembly of network at the surface and resulting retrograde flow (speed va) results in expansion or compression of the intervening network, thereby generating a stress on the nucleus surface. B) Key components of the model for a spreading cell. The model cell accounts for (i) resistance of the nucleus to volume expansion /compression; ii) resistance of the nuclear surface (lamina) to area expansion; iii) cell membrane tension; and iv) the cytoskeletal network phase of the cytoplasm, which is assembled at the cell cortex and at the contact boundary with the substratum. Centripetal flow of network and the frictional resistance to shear and to volume expansion/compression causes movement of the cell membrane and nuclear surfaces (surface velocities of cell and nuclear surfaces are shown by blue vectors). C) Snapshots from a simulation of cell spreading and nuclear shape changes showing the three phases of nuclear deformation observed experimentally: i) vertical distension and translation of the nucleus toward the substratum, driven by flow of network from the apical membrane where it is generated, ii) initial flattening against the substratum with a decrease in nuclear height and little change in nuclear width as the cell begins to spread and the nuclear is compressed vertically by the lowering upper cell surface; and iii) widening of the nucleus with lesser change in height, as the widening cell boundary pulls the nucleus laterally. Widths and height are plotted in D). As in the experimental observation, the nucleus quickly flattens vertically early in the process of cell spreading, then widens more slowly as the cell continues to spread. E) Plot of nuclear area and volume versus time. While the nuclear volume remains nearly constant, the area expands until the assumed excess area is smoothed and the stiffer “true” surface area is reached, at which point the surface area starts to level off toward a constant value.

Page 68: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

68

Page 69: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

69

Figure 2-20. Snapshots of simulation results for different parameters. A). Simulation for case of no cytomatrix assembly at the cortex (assembly occurs only at the contact boundary). Network assembly at the cortex is required for translation toward the substratum. B). Simulation showing the effect of reduced adhesion. Reducing adhesion allows retrograde flow at the substratum; ultimately the flow speed matches the speed of network assembly, yielding steady-state cell and nucleus shapes. The nucleus shape changes cease when spreading stops. C). Simulation with no contractile stress within the network (σc = 0)). Nuclear flattening is predicted to arise from flow alone without requiring actomyosin contractile stresses in the network, consistent with the observation of flattening under myosin inhibition. D). Simulation of a cell with no resistance to nuclear lamina area expansion. Without area stiffness, the nucleus continues to flatten at a nearly constant volume. E). Plot of nuclear height (blue line), width (black line), and cell spreading area (green line) versus time for the cases show in A-D. F). Plot of apparent nuclear surface area (blue line) and volume (green line) for cases A-E.

Page 70: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

70

Page 71: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

71

CHAPTER 3 DYNAMIC DEFORMATION OF THE CELL PLASTICALLY SHAPES THE NUCLEUS

AND AMPLIFIES CANCER NUCLEAR IRREGULARITIES

The aberrant morphology of nuclei is a distinct feature observed in diverse types

of cancer [63-65] used for diagnosis and prognosis [66, 67]. These features include

lobes, invaginations and folds in the nuclear lamina. Given the physical linkage between

the nuclear lamina and chromatin [68, 69], such alterations in nuclear shape can affect

chromatin compaction and consequently gene expression [17, 70-72], causing

alterations of cellular functions such as migration [73] and mitosis [74, 75].

The mechanisms for abnormal shapes of cancer nuclei have remained unclear.

Some papers have suggested that chromosomal instabilities correlate with nuclear

shape abnormalities [74, 76] in cancer cells. Other studies suggest that spatial

inhomogeneity in the nuclear lamins contribute to abnormal cancer nuclear shapes [77].

Based on the new mechanism identified in chapter 2, here we hypothesized that cancer

nuclear abnormalities are caused by stresses generated by the dynamic process of cell

spreading. To test this hypothesis, we performed 3D confocal imaging of cancer nuclear

shapes during the process of cell spreading. Using a convex hull method to quantify

three dimensional nuclear shape abnormalities, we found that nuclear abnormality is

amplified during the dynamic process of cell spreading in breast cancer cells. The

nuclear deformation driven by cell shape changes are also irreversible as no elastic

Reprinted from Journal of Cellular Physiology, in press, Tocco, V., Li, Y., Christopher, K., Matthews, J., Aggarwal, V., Paschall, L., Luesch, H., Licht, J., Dickinson, R., and Lele, T., The nucleus is irreversibly shaped by motion of cell boundaries in cancer and non-cancer cells. Copyright (2017), with permission from Wiley.

Page 72: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

72

relaxation of nucleus is observed after isolating nucleus from cytoplasm in fibroblasts

and cancer cells.

As the LINC complex transmits mechanical stresses generated by cell spreading

to the nuclear surface, we next asked if abnormal nuclear shapes can be ‘reverted’ to

normal shapes by disrupting the LINC complex. Disrupting the LINC complex by

overexpression of GFP-Sun1L-KDEL or GFP-KASH4 caused a decrease in the nuclear

shape abnormalities in MDA-MB-231 cells. Importantly, the motility of breast cancer

cells decreased significantly upon LINC disruption. We further quantified the amount of

DNA in cancer cells using a high-content imaging method. Upon comparing MCF10A

and MDA-MB-231 cells, we found no significant differences between the dependence of

nuclear shape abnormalities on DNA content. However, we found that abnormalities in

nuclear shape are heritable from mother to daughter breast cancer cells.

Taken together, our results are consistent with a model of cancer nuclear

shaping in which motion of the cell boundary transmits a viscous (dissipative) force on

the nuclear surface to amplify irregularities of nuclear shape.

Materials and Methods

Cell Culture and Transfection

All cells were maintained in a humidified incubator at 37°C and 7% CO2. Human

breast cancer cells (MDA-MB-231) were cultured in 4.5g/L glucose Dulbecco’s modified

Eagle’s medium (DMEM) without HEPES and L-glutamine (Invitrogen, Carlsbad, CA),

supplemented with 10% (v/v) donor bovine serum (DBS, Gibco, Grand Island, NY), 1%

(v/v) 100x MEM non-essential amino acid (Mediatech, Manassas, VA), 1% (v/v) 200mM

L-glutamine (Fisher Scientific, Hampton, NH) and 1% Penicillin Streptomycin

(Mediatech, Manassas, VA). Human breast epithelial cells (MCF-10A) were cultured in

Page 73: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

73

DMEM/F12 (Invitrogen, Carlsbad, CA), supplemented with 5% (v/v) horse serum

(Invitrogen, Carlsbad, CA), 1% Penicillin Streptomycin (Mediatech, Manassas, VA), 20

ng/mL epidermal growth factor (EGF, Peprotech, Rocky Hill, NJ), 0.5 µg/mL

hydrocortisone (Sigma-Aldrich, St. Louis, MO), 100 ng/mL cholera toxin (Sigma-Aldrich,

St. Louis, MO) and 10 µg/mL insulin (Sigma-Aldrich, St. Louis, MO). Cells were seeded

onto 35 mm glass-bottom dishes (WPI, Sarasota, FL) treated with 5 µg/ml fibronectin

(BD Biosciences, San Jose, CA) for imaging. Transfections of were performed with

Lipofectamine 3000 (ThermoFisher Scientific, Waltham, MA) in OptiMEM serum-free

media (ThermoFisher) following the manufacturer’s protocols

Cell Staining and Drug Treatment

For fixed-cell experiments, cells were fixed in 4% paraformaldehyde at room

temperature (25°C) for 10 min, washed with PBS, and stained with Hoechst 33342 and

fluorescent phalloidin to label DNA and F-actin, respectively. For microtubule

immunostaining, cells were first treated with microtubule extraction buffer containing

0.5% (w/v) glutaraldehyde, 0.8% formaldehyde and 0.5% Triton X-100 in phosphate

buffered saline (PBS) for 3 minutes before fixing with 1% (w/v) paraformaldehyde for

another 10 minutes. Then a freshly prepared 1% (w/v) sodium borohydride in PBS

solution was added to the cells for 10 minutes followed by blocking in 1% (w/v) BSA in

PBS. The cells were then incubated in 4°C overnight with rabbit polyclonal antibody to

α-tubulin (1:1000, Abcam, Cambridge, MA) in 1% BSA-containing solution, washed with

PBS and then incubated with Goat Anti-Rabbit IgG (H+L) antibody (1:500, Life

Technologies, Carlsbad, CA) at room temperature for 1 hour. For treatment with specific

drug, cells were pretreated for 1hr and imaged in the media containing the drug with the

same concentration

Page 74: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

74

Nuclear Excision

The work was performed by Keith Christopher. In short, the nucleus was

removed from MDA-MB-231 cells by using a 0.5 m micropipette tip (Femtotip;

Eppendorf North America, Hauppauge, NY) as a chisel. The micropipette tip was

controlled with an Eppendorf InjectMan micromanipulator system (Eppendorf North

America, Hauppauge, NY). The change of nuclear shape was recorded before and after

nuclear isolation.

Cell Spreading Assay

The seeding experiments were performed as described in the paper [78]. In brief,

cells were trypsinized and seeded onto FN coated glass-bottomed dishes in the

environment of 37°C and 5% CO2 before fixation with paraformaldehyde at different

time. In the dynamic study, cell was maintained in an environmental chamber with 37 C,

5% CO2 and imaged up to 1hr.

Imaging and Image Analysis

Imaging was done using a Nikon A1 laser scanning confocal microscope (Nikon,

Melville, NY) with a 60x/1.4NA oil immersion objective. For live cell imaging, cells were

maintained at 37 °C and 5% CO2 in a humidified chamber. Z-stacks were acquired with

a 0.3 µm axial step size for fixed cell imaging, and 1–2 µm axial step size for live cell

imaging. Fiji software [79] was used for image processing and all measurements.

For nuclear measurements, the nuclear outline was either traced by hand (for

experiments without fluorescence) or the nuclear parameters determined automatically

from applying an intensity threshold to maximum intensity XY nuclear projections of

fluorescent nuclei. Nuclear contour ratio of cancer cells was calculated as 4πA/P2,

where A is the area and P is the perimeter of the maximum intensity projection.

Page 75: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

75

To measure the three-dimensional abnormality, z-stacks were taken at an

interval of 0.3 and 0.75 µm, for fixed and live cells respectively, and X-Y position of each

pixel along the nuclear edge in each z-stack was detected with threshold determined by

Ostu method and recorded with Image J (NIH). Meanwhile, the nuclear volume was

measured with same threshold by Image J (NIH). The coordinate data of pixels in all z-

stacks were imported into Python software to calculate the volume of the convex hull

covering the whole nucleus with its integrated convex hull function. The three-

dimensional abnormality is calculated by dividing the difference between measured

nuclear volume and convex hull volume with measured nuclear volume. The intensity of

GFP-NLS signal in nucleus and cytoplasm was measured by Image J (NIH).

High-Content Imaging

Both MDA-MB-231 and MCF-10A cells were seeded on 384-well plates and then

fixed at approximately 95% confluence. The nuclei were stained with same

concentration of H33342 dye and imaged with an Operetta CLS system (PerkinElmer,

Waltham, MA) with the same intensity of UV light. The images were analyzed by Cell

Profiler software (Carpenter lab, Boston, MA) to acquire data of nuclear morphology and

signal intensity.

Results

The Deforming Cell Shape Amplifies Cancer Nuclear Abnormalities

To gain insight into how irregular cancer nuclear shapes are established, we

quantitatively tracked nuclear irregularities during cell spreading by imaging GFP-NLS

expressing nuclei in MCF-10A and MDA-MB-231 cells (Figure 3-1A). We observed

instances of transient nuclear membrane rupture (indicated by a loss and later recovery

of GFP-NLS signal in the nucleus) during spreading in MDA-MB-231 cells but not in

Page 76: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

76

MCF-10A cells (Figure 3-1A). The occurrence of membrane rupture indicates the

presence of mechanical stresses on the nuclear membranes during spreading. To

calculate irregularities while the nucleus deforms significantly in three dimensions, we fit

a convex hull to the three-dimensional nuclear shape constructed from confocal stacks,

and calculated the fractional deviation of nuclear volume from the fitted hull. The 3-D

abnormality increased in cancer nuclei dynamically during spreading of cancer cells, but

not in MCF-10A cells (Figure 3-1B). These results show that dynamic deformation of the

cell during spreading amplifies nuclear irregularities in cancer cells, but not in non-

malignant cells. Consistent with our previous results, cancer nuclear abnormality

increases during spreading even in the absence of actomyosin contraction, although the

increase is attenuated (Figure 3-1C). We therefore cannot rule out a contribution of

actomyosin stresses in establishing abnormal nuclear shapes.

Disrupting Either the LINC Complex or the Cytoskeleton Dampens Cancer Nuclear Abnormality

Cytoskeletal forces are transmitted to the nuclear lamina and associated proteins

through nuclear envelope proteins that form the Linker of the Nucleus to Cytoskeleton

(LINC) complex [34, 80, 81]. We therefore hypothesized that LINC complex disruption in

MDA-MB-231 cells normalizes the nuclear shape. We disrupted the LINC complex with

two approaches: overexpression of GFP-KASH4 and GFP-SUN1L-KDEL. KASH4 is the

binding domain of nesprin4 interacting with SUN1/2 in the inner nuclear membrane, and

overexpression of GFP-KASH4 competitively prevents the binding of endogenous

nepsrin proteins with SUN1/2, which consequently disrupts the LINC complex. GFP-

SUN1L-KDEL is negative domain protein disabled to bind with KASH domain, which

can disconnect nuclear membrane and cytoskeleton by competing with their

Page 77: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

77

endogenous counterparts after overexpression. LINC complex disruption with either

KASH4 or SUN1L-KDEL over-expression indeed reduced the three-dimensional

abnormality of the nucleus without change of cell spreading area (Figure3-2B and C).

Consistent with this data, disrupting cytoskeletal forces also reduced the 3D abnormality

of the nucleus (Figure 3-3A and B) with little change of cell spreading area (Figure 3-3

C), either through myosin inhibition by blebbistatin or disrupting microtubules with

nocodazole treatment. Conversely, overexpressing laminB1 or laminA/C both increased

3D nuclear abnormality (Figure 3-2B).

The Reduction of Nuclear Abnormality by LINC Complex Disruption Impairs Cellular Motility

As shown above, LINC disruption ‘normalized’ the abnormal cancer nuclear

shape. Based on previous results from Alam et al [37] which showed that transmission

of forces to the nucleus is necessary for persistent cell migration, we hypothesized that

LINC disruption in cancer cells reduces cancer cell migration. The LINC complex was

again disrupted with GFP-KASH or GFP-SUN1L-KDEL expression as above. Compared

with control groups (GFP and GFP-KDEL), migration is significantly reduced in LINC-

disrupted cells (GFP-KASH4 and GFP-SUN1L-KDEL, Figure 3-4). This is reflected in

over 50% drop in the mean squared displacement (MSD) (Figure 3-5). This drop in

migration is likely due to the fact that the nucleus does not transmit forces efficiently,

because recent data from the Lele lab suggests that cytoskeletal signaling pathways

that control migration are unaltered in LINC disrupted cells (data not shown).

Abnormal Morphology of Cancer Nucleus do NOT Necessarily Reflect Chromatin Content

There is some evidence that chromatin instabilities could cause abnormal

nuclear shapes in cancer [74, 76]. We showed above that nuclear abnormalities

Page 78: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

78

increase during cancer cell spreading (as opposed to decreasing in normal cell

spreading). Here we asked if the ploidy (chromosomal content) in cancer nuclei

correlates with nuclear shape abnormalities. To do this, we imaged a large number of

nuclei stained with H33342 with a high content imaging technique. We observed a

bimodal distribution of the total amount of DNA content in both MCF-10A and MDA-MB-

231 cells, consistent with cells being in different phases of the cell cycle (Figure 3-6A).

MDA-MB-231 cells were observed to have a long tail in the distribution, suggestive of

aneuploidy (Figure 3-6A). MDA-MB-231 cells had higher average DNA content as

evident from the right-shifted distribution of MDA-MB-231 cells compared with MCF-10A

cells (Figure 3-6B). We found only minor difference in the dependence of 2-D nuclear

abnormality on DNA content (Figure 3-6C) between MDA-MB-231 with MCF-10A cells.

DNA content also did not correlate with 3-D nuclear abnormality (Figure 3-6D). Taken

together, these results suggest that abnormal nuclear morphology is not necessarily

reflective of chromatin content.

Nuclear Abnormality can be Inherited by Offspring

We next examined if cells containing abnormal nuclear shapes have defects in

proliferation. MDA-MB-231 cells with either normal or abnormal nucleus were imaged

up to 18 hr. In this time period, cells with abnormally shaped nuclei were half as likely to

divide as cells that contained a normal nucleus (Figure 3-7A). We also tracked nuclear

shapes in daughter cells. Mother cells with regular nuclei tended to divide into cells with

regular nuclei, while cells with abnormal nuclei (Figure 3-7B) tended to divide into cells

with abnormal nuclei. We also found that over half of the cells with highly abnormal

nuclei (nuclei with two or more lobed structures) were able to remain alive up to 18hr

imaging and the fraction of cell with successful division is similar with cells with regularly

Page 79: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

79

abnormal nuclei (Figure 3-7C). These results indicate that the abnormal shape of the

nucleus attenuates but does not eliminate proliferation in breast cancer cells.

Discussion

Nuclear Abnormalities in Cancer.

We quantified the degree of abnormality in the cancer nuclear shape during

dynamic deformations in cell shape. The abnormality increased in MDA-MB-231 cells,

but not in MCF-10A cells during the process of cell spreading. Coupled with the

irreversibility of the shape deformations upon cell microdissection and the observed

reduction of cancer nuclear irregularities upon LINC disruption, we interpret these

results to suggest that mechanical stress on the cancer nucleus generated by the

dynamic process of spreading amplifies cancer nuclear irregularities. Furthermore,

treatment with Y27632 which inhibits myosin but promotes actin polymerization, did not

prevent the increase in the nuclear abnormalities during cell spreading. This is similar to

our observations in Chapter 2 where myosin activity was not required for nuclear

shaping during cell spreading.

Recent literature suggests that many LINC complex components are

downregulated, both in patient breast tumors and breast cancer cell lines [14]. SUN1

and SUN2 levels are downregulated in breast cancer tumors relative to surrounding

non-cancer tissue, and genes encoding nesprin-1 and nesprin-2 are mutated in breast

cancer tissue [82, 83]. Our result that LINC disruption reduces cancer nuclear

irregularities suggests that the LINC complex is still mechanically functional in cancer

cells despite such changes to its protein components, and contributes to nuclear

irregularities.

Page 80: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

80

It has been reported that lamins in cancer nuclei are spatially inhomogeneously

distributed in cancer nuclei resulting in a mechanically ‘patchy’ nuclear surface [77].

Thus, certain regions of the heterogeneous nuclear surface may be more compliant in

response to mechanical stresses, resulting in the observed irregular nuclear shapes.

We note here that knockdown of nuclear proteins other than the nuclear lamins [84] can

also result in irregularities- these include the chromatin remodeling protein BRG1 [85]

and endosome regulator protein Wash [86].

We have previously shown that cell spreading is necessary and sufficient to drive

nuclear flattening in fibroblasts under a wide range of conditions [78], including in the

absence of microtubules, vimentin intermediate filaments and myosin activity.

Therefore, we speculate that here it is primarily the F-actin network that transmits stress

from the moving membrane to the cancer nucleus. However, once F-actin is completely

disrupted with pharmacological agents, any further cell protrusion and spreading are not

possible [78], which makes it difficult to test the role of F-actin in mediating nuclear

response to deformations in cell shape over the required time scale, at least several

minutes. Alternatively, it is possible that any of the three cytoskeletal filaments can

transmit stresses to the nucleus, and disruption of one of these initiates other filaments

to engage in the force transmission. If there is such redundancy, then identifying the

molecular structures that cause an increase in cancer nuclear abnormalities is likely to

remain a fundamental challenge.

The shape of the nucleus has been commonly assumed to store elastic energy.

The prediction of this model is that nucleus should restore the original rounded shape

once removing cytoplasmic forces. Similarly, nuclear lobes and invaginations,

Page 81: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

81

characteristic of cancer nuclei, are thought to be elastically deformed in response to

cytoskeletal forces. These results are contradicted, however, by recent results from the

Lele lab [87] that isolation of the cancer nucleus from the cell by micro-dissection does

not cause a relaxation of the nuclear shape. The lack of elastic relaxation upon removal

of the surrounding cell implies that the deformed nuclear shape in cancer cells stores no

elastic energy and that the deformation is not an elastic response to the instantaneous

cell shape-dependent cytoskeletal forces on the nucleus.

Because gene expression and protein synthesis potentially depend on the

physical properties of the nucleus [17, 55, 88], reversible deformations of the cancer

nucleus may trigger expression of genes during cell migration by modulating chromatin

compaction [89, 90]. Therefore, knowing how dynamic cell shape information is

transmitted to the cancer nucleus will be key to a complete understanding of the

relationship between cell shape and cell function.

Mechanical Stress and Cancer Cell Migration.

Our observation that LINC disruption normalizes the nucleus and reduces cancer

cell motility is surprising considering that the LINC complex components are

downregulated in MDA-MB-231 cells [14]. It appears that the LINC complex continues

to be functional in these cells despite low levels of these proteins. Unpublished results

from the Lele lab have shown that LINC disruption does not alter Rho/Rac/Cdc42

signaling pathways in these cells. Wu et al. [57] and Alam et al. [37] have suggested

that the nucleus can act as an intracellular scaffold, transmitting forces from one end of

the cell to the other. This mechanical integration allows the cell to migrate efficiently.

Thus, we interpret these results to suggest that an intact LINC complex transmits forces

in breast cancer cells.

Page 82: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

82

The existence of aneuploidy or an abnormal number of chromosomes has been

shown in most cancer types and is known to promote the progression of cancer [76, 91,

92]. As expected, our results show that MDA-MB-231 cells have more DNA than

MCF10A cells. Coupled with the fact that MDA-MB-231 cells have abnormal nuclei, one

might conclude that abnormal shapes of nuclei correlate with DNA content. However,

further examination shows that there is little correlation between DNA content and

nuclear shape abnormalities. Therefore, we conclude that nuclear shape abnormalities

in cultured cancer cells are not explained by DNA content alone. We have also shown

that the abnormality of nuclear shapes is heritable. The mechanism for this is unclear

and will need to be probed further in future studies.

Page 83: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

83

Figure 3-1. The abnormality of nucleus amplifies during cell spreading. A) Shown is an

example of nuclear rupture evident from the loss of GFP-NLS from the nucleus of a spreading MDA-MB-231 cell. An MCF-10A cell during spreading is shown for comparison. The change of NLS signal intensity normalized with its initial intensity is shown in line plots at right of the corresponding cell type. Scale bar is 10µm B) Schematic shows the calculation of 3D abnormality defined here as the difference in volume between the nucleus and a convex hull fit to the nucleus divided by nuclear volume. Plot shows 3D abnormality plotted against time of spreading in MCF-10A and MDA-MB-231 cells. C) The abnormality of nuclear shape increases during cell spreading even in the absence of myosin contraction. (n>30 at each time point, data are shown as Mean ± SEM, p value of ANOVA analysis is 0.02, 0.04 and 0.83 for DMSO, Y27 and Blebbistatin, respectively)

Page 84: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

84

Page 85: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

85

Figure 3-2. Disrupting the LINC complex reduces nuclear abnormality. A). Images of nuclei stained with H33342 (blue) are shown in cells transfected with GFP (control), GFP-KASH4, GFP-KDEL (control), GFP-SUN1L-KDEL, GFP-Lamin A/C or GFP-lamin B1. The 3-D nuclear abnormality B) and cell spreading area C) for populations of cells quantified. (Three biological replicates with n=59, 62, 90, 86, 93 and 72 for 3D nuclear abnormality of GFP, GFP-KASH4, GFP-KDEL, GFP-SUN1L-KDEL, GFP-Lamin A, GFP-Lamin B1, respectively; and n>30 for cell spreading area of all conditions. Data are shown as Mean ± SEM, * indicates p<0.05 by t-test)

Page 86: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

86

Figure 3-3. Disrupting cytoskeletal elements reduces nuclear abnormality. A) Images of cells treated with latrunculin A (Lat A), blebbistatin (Blebb), or nocodazole (Noco) are shown stained for microtubules (green), actin (red) and DNA (blue). The 3-D nuclear abnormality B) and cell spreading area C) for populations of cells quantified.(Three biological replicates with n=39, 142, 107 and 75 of nuclear 3D abnormality for wild type, nocodazole, blebbistatin, and latrunculin A, respectively; and n>30 for cell spreading area of all conditions. Data are shown as Mean ± SEM, * indicates p<0.05 by t-test).

Page 87: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

87

Figure 3-4. Trajectory maps of MDA-MB-231 cells with or without LINC disruption.

Each color line in the trajectory maps represent one cell (More than Three biological replicates with n=42, 40, 40 and 39 for GFP, GFP-KASH4, GFP-KDEL and GFP-SUN1L-KDEL, respectively). The inserted images at top right corner of trajectory map are the representative images of transfected cell for each condition.

Page 88: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

88

Figure 3-5. LINC complex disruption impairs cellular motility. Shown is the MSD of individual cell (A) and plot of average MSD (B) in all four conditions. (More than Three biological replicates with n=42, 40, 40 and 39 for GFP, GFP-KASH4, GFP-KDEL and GFP-SUN1L-KDEL, respectively. Data are shown as Mean ± SEM, * indicates p<0.05 by Mann-Whitney Test).

Page 89: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

89

Figure 3-6. The abnormal shape of nucleus does not correlate with DNA content. A) Shown is the distribution of total DNA content (proportional to the total signal intensity of H33342) of MCF-10A and MDA-MB-231 cells (Three biological replicates with n = 9995 and 7970 for MCF-10A and MDA-MB-231, respectively). B) Shown is the distribution of average DNA content (defined as total H33342 intensity over total pixel number) for MCF-10A and MDA-MB-231 cells. C) Nuclei of MCF-10A and MDA-MB-231 cells were binned according to total DNA content and the average nuclear 2D abnormality (defined as the difference in nuclear area between the nucleus and a convex hull fit to the nucleus divided by nuclear area) is plotted for each bin. Error bars represent SEM for n = 625, 7157, 1633, 494, 81 (MCF-10A) and 0, 232, 2839, 2970, 1865 (MDA-MB-231) nuclei for each bin, respectively. D) Scatter plot of total DNA content with respect to nuclear 3D abnormality. (Data are shown as Mean ± SEM, * indicates p<0.05 by t-test).

Page 90: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

90

Page 91: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

91

Figure 3-7 The abnormality of nuclear shape is heritable. A) MDA-MB-231 cells were manually classified as having a regular or irregular nucleus (More than Three biological replicates with n = 225 and 281, respectively) and tracked for 18 hours with DIC microscopy. As shown, 14% of cells with regular nuclei and 6% of cells with irregular nuclei underwent mitosis during this period. B) Of the cells that divided, the resulting daughter nuclei were classified as regular and irregular (as before) and plotted in the shown pie charts (“unclear” indicates that the nucleus could not be classified due to changing focal plane or migrating out of the field of view). C) Shown is the pie plot about the fate of mother cell with highly irregular nuclei (“highly” is identified according to the number of lobules of nucleus, which is at least two).

Page 92: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

92

CHAPTER 4 CONCLUSIONS

Summary of Findings

This thesis has discovered a novel mechanism for nuclear shaping. We

discovered that the degree of nuclear flattening is closely tied to the degree of cell

spreading. For example, Figure 4.1 shows a plot of the x-z nuclear aspect ratio versus

cell spreading area. This data is pooled from fibroblast experiments across different

conditions- myosin inhibition, microtubule disruption, what else, etc. As seen, all data

seem to suggest a ‘master’ curve that captures the fact that nuclear flattening is

inversely correlated with the degree of spreading. As long as cells are able to spread,

the nucleus is flattened. This observation directly challenges the currently accepted

model in the literature in which myosin activity causes actomyosin bundles to compress

and thereby shape the nucleus [22, 38].

To explain the dependence of nuclear flattening on cell spreading, we have

proposed a model in which motion of the cell boundary transmits a frictional stress

through the cytoskeleton to the nuclear surface. Model calculations show that this

mechanism adequately explains how the vertical nuclear shape mimics vertical cell

shape. After publication of our paper, Alam et al. [37] reported that local protrusions

proximal to the nuclear surface can transiently deform the nucleus, and relaxation of the

protrusions caused relaxation of the deformed shape (Figure 4.2). Similarly, Neelam et

al. [93] have shown that vertical nuclear shapes mimic vertical cell shapes in confluent

epithelial cell monolayers. Our proposed mechanism in which the nuclear shape mimics

cell shape through local movement of cell boundaries can easily explain how the

nucleus takes on an hour-glass shape in cells migrating through small pores as

Page 93: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

93

observed by Lammerding, Friedl and coworkers [94, 95]. In contrast, it is not clear how

actomyosin bundles could compress the nucleus into such hourglass shapes that mimic

cell shapes.

Our work also suggested that the nucleus reaches a steady-state shape once the

nuclear lamina becomes completely taut, beyond which is stiff to cellular forces. Our

concept has since received support from direct measurements of the stiffness of

isolated nuclei by Stephens et al. [96]. They showed that the nucleus is soft below

~30% strains and this stiffness is due to chromatin inside the nucleus. Beyond this

strain, the stiffness increases abruptly and the authors attribute this to the taut nuclear

lamina. Also, Neelam et al. [93] have since shown that the folds in the nuclear lamina

decrease with cell spreading in MCF10A cells.

During cell spreading, we showed that the nuclear volume remains constant. Our

results thus suggest a mechanistic picture in which nuclear shape changes in the cell at

constant volume and constant surface area. Lammerding and coworkers have also

shown that the nuclear volume does not change during the large deformations observed

of the nucleus as the cell migrates through narrow pores [97].

If nuclear volume and surface area remain constant during changes in nuclear

shape, then relieving stresses on the nuclear surface should produce no changes in

nuclear shape. This concept directly contradicts the notion that the nucleus is elastically

deformed in cells, an assumption made in all current models in the literature [98-101].

Our concept has since received support from experiments by Keith Christopher in which

he removed nucleus from cell body by micro-excision and showed that the nucleus

Page 94: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

94

undergoes no change in its shape upon isolation from the cell [87]. No elastic energy is

stored in the nucleus inside cells, both for fibroblasts as well as cancer cells.

We also demonstrated that lobes and invaginations in the cancer nucleus are

amplified by the process of cell spreading, and this amplification again does not require

myosin activity. This is not to suggest that actomyosin bundles cannot indent the

nuclear surface or constrict it in cancer cells after the initial nuclear shape is established

[102]. However, the frictional stress transmission is responsible for establishing the

steady-state shapes during processes like cell spreading.

Future Work

We proposed that the cell exerts frictional or viscous stresses on the nuclear

surface based on the physical concept that the cellular cytoskeleton does not store

elastic stresses over the time scales of several minutes over which the nuclear and cell

shapes are established. However, direct evidence that the stresses are frictional in

nature has not been developed yet. Experiments that correlate the rate of motion of the

cell boundary with the rate of motion of the nuclear surface could offer stronger support

to this transmission mechanism.

A crucial open question for the mechanism proposed in this thesis is the identity

of the cytoskeletal elements that propagate the frictional stresses to the nuclear surface.

We have already shown that microtubules, intermediate filaments and myosin activity

are not required for this stress transmission mechanism. We therefore hypothesize that

F-actin structures transmit these frictional stresses to shape the nucleus. This

hypothesis is challenging to test because without changes in cell shape, the nucleus

cannot change shape, and disrupting F-actin filaments rounds up the cell.

Page 95: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

95

One possible experiment is to test the hypothesis that proteins which cross-link

F-actin filaments to transmit stresses. Such cross-linking proteins include alpha-actinin

and filamin. Assuming that knockdown of these proteins does not prevent F-actin

polymerization, it is possible that the nucleus will remain ‘rounded’ during cell spreading

and therefore prevent cell spreading despite normal F-actin polymerization at the

leading edge.

It is also possible that all three cytoskeletal elements, as well as membranous

structures like the endoplasmic reticulum or the Golgi apparatus might transmit stresses

to the nuclear surface. Even though disruption of the microtubules or intermediate

filaments does not prevent nuclear flattening, it is possible that these structures act

redundantly to transmit stresses. If this is indeed the case, then it will remain difficult to

unequivocally identify the structures that participate in the shaping of the nucleus.

How the nucleus itself behaves during shaping is also of interest. Our data

suggests that inhibiting myosin activity reduces nuclear volume. The nuclear volume

resists changes presumably because of osmotic stresses exerted by macromolecules

that do not leave the nucleus. This property likely accounts for the resistance to volume

expansion or compression. Therefore it is possible that myosin inhibition decreases cell

volume, which causes osmotic flow out of the nucleus and thereby reduce its volume.

Whether this mechanism is indeed responsible for decrease in the nuclear volume

remains unknown. Experiments which measure cell volume simultaneously with myosin

inhibition can prove useful. However, cell volume is not easy to accurately measure in

spread cells with current methods including confocal microscopy. Other measurements

Page 96: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

96

might involve quantifying the concentration of tracer molecules upon myosin inhibition in

the cytoplasm and the nucleus.

We have shown that cancer nuclear abnormalities increase during spreading.

The reasons for this are unclear. It is possible that the nucleus has a mechanically

patchy surface as has been suggested by others [77], but direct evidence is not

available for this. It may be possible that nuclear manipulation experiments developed

by Neelam et al. [103] might be used to reveal if cancer nuclei indeed have spatially

distributed mechanical properties in the same cancer nucleus.

Our assays have focused on in vitro model systems in which cells spread on 2-D

surfaces. Whether these principles apply in tissues inside organisms remains to be

established. Nuclei in tissues formed by epithelial cells do not have ‘flattened’ nuclei.

Cells in epithelia tend to be cuboidal with corresponding nuclear shapes. We predict

that the mechanism by which these nuclear shapes are established in these tissues is

similar to the mechanisms we have discovered with in vitro model systems. While

testing these mechanisms in vivo is highly challenging, we expect that progress will be

made with intermediate model systems, such as 3D mammary epithelial acinar

assemblies in matrigel.

Page 97: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

97

Figure 4-1 Data pool of x-z nuclear aspect ratio versus cell spreading area. (Three

biological replicates with n=40, 34, 43, 38, 39, 35, 30, 33 and 34 for Control, ML-7, Y27, Blebbistatin, Nocodazole0.83µM, Vim+/+, Vim-/- MEF, Nocodazole1.67µM and Blebbistatin 6hr, respectively).

Page 98: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

98

Courtesy of Samer Alam [37] Figure 4-2 Local nuclear deformation in response to local protrusion and retraction of

cell memrbane. Shown is the formation and retraction of a lateral protrusion of cell membrane near the nucleus at various confocal planes (planes 1-4 in cartoon on the left), and the reversible nuclear deformation (in the xy-plane) accompanying the protrusion. YZ view of the nucleus at the mid-plane shows the deformation in the yz-plane. Vertical dashed lines indicate the position of the corresponding yz plane. Cell is labeled with RFP-LifeAct for F-actin and GFP-histone H1 for nucleus. Scale bar, 10μm. Photo credits Samer Alam.

Page 99: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

99

APPENDIX A COMPUTATIONAL MODEL FOR NUCLEAR DEFORMATION DURING CELL

SPREADING

Constitutive Model for Cytoskeletal Network Stress

The assumed constitutive equation for the stress tensor in the network phase of

the cytoplasm is

𝛔 = 2𝜇�̇� + (σ𝐜 + λ∇ ∙ 𝐯)𝐈 A1-1

where I is the unit dyadic, �̇� = 1

2(∇𝐯 + ∇𝐯𝑻) is the rate-of-strain tensor, and 𝜇 and λ are

viscosity parameters. Eq. A1-1 models the cytoskeletal network as a compressible

contractile network. Network density changes, which may affect these properties, are

assumed to equilibrate by local assembly/disassembly over the slow time-scale of cell

spreading; therefore no continuity equation for the network density is required. Since

network volume is not locally conserved, Eq. A1-1 reflects both shear and

expansion/compression strains. If the strains caused by both modes of deformation

have the equivalent resistances, then we can assume λ ~0, reducing Eq. A1-1 to a

single viscosity parameter 𝜇. (In linear elasticity, this is analogous to assuming

Poisson’s ratio is zero such that the Young’s modulus (a measure of longitudinal

stiffness) equates to twice the shear modulus (a measure of shear stiffness).

The longitudinal transmission of normal stress to a surface due to a distant

moving boundary is an important property of Eq. 1.1 that is relevant to our model for cell

spreading. To illustrate this, consider first a one-dimensional case of a contracting

network that is fixed at one end (𝑣𝑥(𝑥 = 0) = 0) and moving with velocity V at a distance

𝑥 = 𝐿 (i.e. 𝑣𝑥(𝑥 = 𝐿) = 𝑉)). The stress balance ∇ ∙ 𝛔 = 𝟎 in the x-direction is

𝑑𝜎𝑥𝑥

𝑑𝑥= 2𝜇

𝑑2𝑣𝑥

𝑑𝑥2 = 0 A1-2

Page 100: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

100

Applying the boundary conditions yields the velocity field

𝑣𝑥(𝑥) = 𝑉𝑥/𝐿 A1-3

as well as the stress field

𝜎𝑥𝑥 = σ𝐜 + 2𝜇𝑉/𝐿 A1-4

which is uniform in this case. Hence, noting the second term in Eq. A1-4, translating the

one boundary at 𝑥 = 𝐿 at speed V transmits an additional stress 2𝜇𝑉/𝐿 to the surface at

𝑥 = 0 due to longitudinal friction, which is positive for expansion (𝑉 > 0), and negative

for compression (𝑉 < 0).

Now consider a spherical cell of radius R with a nucleus of radius 𝑅𝑛, under the

assumption of spherical symmetry, the stress balance in the cytoplasm is

𝑑𝜎𝑟𝑟

𝑑𝑟+

1

𝑟(2𝜎𝑟𝑟 − 𝜎𝜃𝜃 − 𝜎𝜙𝜙) = 0 A1-5

Where

𝜎𝑟𝑟 = 𝜎𝑐 + 2𝜇𝑑𝑣𝑟

𝑑𝑟

𝜎𝜃𝜃 = 𝜎𝑐 + 2𝜇𝑣𝑟

𝑟 A1-6

𝜎𝜙𝜙 = 𝜎𝑐 + 2𝜇𝑣𝑟

𝑟

Assume now that new network is assembled at the cell membrane and moves

centripetally with speed 𝑣𝑎, and allow the cell radius to expand at speed V (ignoring for

now any volume constraints). Substituting Eqs. 2-2 into Eq. 2-1 and applying the

boundary conditions, 𝑣𝑟(𝑟 = 𝑅𝑛) = 0 and 𝑣𝑟(𝑟 = 𝑅) = 𝑉 − 𝑣𝑎 yields the r-velocity field,

𝑣𝑟 = (𝑉 − 𝑣𝑎)𝑟

𝑅

(1−(𝑅𝑛

𝑟)

3)

(1−(𝑅𝑛𝑅

)3

) A1-7

Eq. A1-3 then provides the rr-component of the stress tensor:

Page 101: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

101

𝜎𝑟𝑟 = 𝜎𝑐 + 2𝜇(𝑉 − 𝑣𝑎)1

𝑅

(1+2(𝑅𝑛

𝑟)

3)

(1−(𝑅𝑛𝑅

)3

) (A1-8)

Such that stress of the nucleus surface is:

𝜎𝑟𝑟(𝑟 = 𝑅𝑛) = 𝜎𝑐 + 2𝜇(𝑉 − 𝑣𝑎)1

𝑅

3

(1−(𝑅𝑛𝑅

)3

) A1-9

(In the small gap limit, 𝐿 = 𝑅 − 𝑅𝑛 ≪ 𝑅 , Eq. A1-7 and A1-8 become equivalent to

Eqs.A1-3 and A1-4.). Therefore, similar to the one-dimensional case, the net tensile

stress on the nucleus is increased by movement of the cell boundary to expand the

network, but is also reduced by assembly of network at the membrane and the resulting

centripetal flow, which causes compression of the intervening network. In this way, the

movement of the cell boundary and network assembly at the cell membrane can

modulate the stresses on the nuclear surface. In general, the nucleus will tend to distort

to follow the changes in cell shape and will follow the flow field generated by network

assembly at the membrane. This is the basis for our model for nuclear shape changes

during cell spreading.

Model for Cell Mechanics

We apply a simple mechanical model of the cell that takes into account (1) the

resistance of the nucleus to volume compression/expansion; (2) resistance of the

nuclear surface to area expansion; (3) tension of the cell membrane, and (4) friction due

to centripetal flow of network tangent to the adhesive substratum. The network normal

stress on the nuclear surface is balanced by the nuclear internal tension 𝜏𝑛𝑢𝑐 (or

pressure when 𝜏𝑛𝑢𝑐 < 0) due to its resistance to volume changes, and the nuclear

surface tension Τ𝑛𝑢𝑐, due to its resistance to surface area expansion. The internal

nuclear tension is modeled as

Page 102: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

102

𝜏𝑛𝑢𝑐 = 𝐾ln(𝑉/𝑉0) A2-1

Where K is the bulk compressibility and 𝑉0 is the unstressed volume. The surface

tension Τ𝑛𝑢𝑐 of the nucleus is expected to depend on strained surface area of the

nuclear lamina A above the unstressed area 𝐴0. We note that surface area undulations

are evident in cross-sectional images of nuclei, indicating roughly 20%-40% excess

area. Therefore, to account for the energy associated with smoothing the nuclear

lamina, we estimated Τ𝑛𝑢𝑐 using the following equation, which is normally applied to

calculate vesicle surface tension accounting for thermal undulations [104]:

𝐴−𝐴0

𝐴0=

𝐸𝑠

8𝜋𝑘𝑐𝑙𝑛 (1 +

𝐴0

24𝜋𝑘𝑐𝑇𝑛𝑢𝑐) +

𝑇𝑛𝑢𝑐

𝜅 A2-2

for > 𝐴0 , where κ is the area extensional modulus of the nuclear lamina, 𝑘𝑐 is its

bending modulus of the lamina, and 𝐸𝑠 is a parameter that can be considered the

magnitude of the energy driving the undulations (equal to 𝑘𝐵𝑇 - Boltzmann’s constant

multiplied by temperature – for undulations driven by thermal energy). In this equation,

the first logarithmic term dominates at low area expansion (low lamina tension), while

the second term dominates at high area expansion. Assuming a value of 𝐸𝑠 ~100 𝑘𝐵𝑇

(Boltzmann’s constant multiplied by temperature) yields excess area in the observed

range, which is reasonable noting intracellular energy fluctuations tend to be on the

order of 100-fold larger that thermal fluctuations [105].

Except for the adhesive substratum, tangential traction stresses on cell and

nuclear membrane surfaces are assumed negligible (i.e., slip boundary conditions).

The normal stress exerted on the cell membrane is assumed to be balanced by the

cell’s internal hydrostatic pressure 𝑃ℎ (assumed uniform throughout the cell and

nucleus) and the stress due to membrane tension 𝑇𝑚𝑒𝑚. Due to the high cytoplasmic

Page 103: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

103

osmolality, cells are resistant to volume changes under typical cellular stresses, hence

simulations were performed under the constraint of constant cell volume, maintained by

varying 𝑃ℎ.

For the boundary at the substratum, network flow at the substratum is assumed

to exert a tangential stress vector equal to 𝜂𝒗(𝑧 = 0), where 𝒗(𝑧 = 0) is the network

velocity tangential to the substratum. The limit 1/η→0 represents the case of perfect

adhesion, such that 𝒗(𝑧 = 0) = 0 (no-slip boundary condition). In either case, it is

assumed there is no network flow in the direction normal to substratum.

To account for cortical actin assembly at the cell membrane, the net boundary

velocity is increased by the actin assembly speed 𝑣𝑎 directed normal to the surface,

except near the substratum contact boundary, where assembly occurs with speed 𝑣𝑎𝑐

directed tangential to the substratum. The net local velocity of the cell membrane is

therefore equal to the difference between the network assembly velocity and the

retrograde flow velocity.

Model Parameters

Parameter Estimates: A list of parameters used in the simulations is shown in

Table A-1. It should be emphasized that key qualitative conclusions from the model –

network flow-driven translation of the nucleus to the surface, nuclear flattening resulting

from cell spreading rather than network tension -- do not strongly depend on several

parameter values, as noted below. Values for nucleus area modulus 𝜅, nuclear bulk

modulus 𝐾 were obtained from measurements by Dahl et al. [11], with the latter

parameter value calculated from their measured osmotic resistance to volume

expansion. Values for membrane tension 𝑇𝑚𝑒𝑚 varies widely from 0.01 nN/μm to 0.3

Page 104: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

104

nN/μm [106, 107], so a mid-range value of 0.1 nN/μm was used (the quantitative

predictions depend only weakly on the value of this parameter). The network assembly

speed at the contact boundary 𝑣𝑎 = 𝑣𝑎𝑐 was estimated from the observed initial speed

of cell spreading (0.5 μm/min). The assembly speed of network at the cell cortex v_a is

not known, but we show results for two cases: 𝑣𝑎 = 𝑣𝑎𝑐 and 𝑣𝑎 = 0, to demonstrate that

cytoskeleton assembly and resulting flow (𝑣𝑎 > 0) is necessary for initial translation and

flattening of the nucleus against the surface. The value of network viscosity was

estimated from the literature [108]. The contractile stress 𝜎𝑐 could be estimated from

Eqs. A1-6 and A2-1, noting that volume was ~50% reduced upon myosin inhibition. If

𝜎𝑐 is assumed to be zero in this case, then 𝜎𝑐 for the control case can be estimated from

the volume difference. Under typical values of other parameters, the second term in Eq.

1.6 is relatively small, such that 𝜎𝑐 ≅ 𝐾 ln(𝑉𝑛 𝑉𝑛∗⁄ ), or 𝜎𝑐 ≅ 0.69𝐾 for a 50% volume

reduction. However, as in the main text, a key prediction is that shape changes during

spreading do not significantly on this background network tension.

Parametric Sensitivity: The key model predictions – translation of the nucleus to

the substratum and flattening of the nucleus during cell spreading – were found depend

to only on the following quantities. The speed of network assembly at the cortex relative

to the contact boundary assembly speed, 𝑣𝑎/𝑣𝑎𝑐 determines how fast the nucleus

translates to the surface while the cell spreads. The nuclear envelope stiffness relative

to viscous stresses (dimensionless ratio 𝜅/𝜇𝑣𝑎𝑐) as well as amount of excess nuclear

surface area (reflected in Eq. A2-2) determines the extent of nucleus flattening in a fully

spread cell. The substratum adhesivity relative to the viscous stresses (dimensionless

ratio 𝜂𝑅𝑛/𝜇) determines the steady-state cell spreading area (hence the amount of

Page 105: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

105

flattening). Model simulations were found to not depend strongly on the bulk modulus of

the nucleus because nuclear shape changes during flattening can proceed without

requiring volume compression (i.e. at constant volume). The contractility parameter 𝜎𝑐

was unimportant since the tension this parameter quantifies was assumed to be uniform

throughout the cytoplasm hence it acts uniformly on all surfaces, and motion is driven

by the divergence of the stress tensor, ∇ ∙ 𝛔 (c.f. Eq A1-1), in which case the constant 𝜎𝑐

disappears. The assumed value of cell membrane tension relative to viscous stresses

(dimensionless ratio, 𝑇𝑚𝑒𝑚𝜇/𝑣𝑎𝑐) had a modest effect on the curvature of the cell

membrane of a spread cell, but had little effect the predicted cell spreading dynamics

and nuclear shape changes.

Methods for Simulating Cell Spreading

The resulting quasistatic stress balance ∇ ∙ 𝛔 = 0 based on Eq. A1-1 is

mathematically equivalent to the classic problem of elastostatic deformation of an

isotropic elastic medium. Therefore, axisymmetric velocity field 𝑣𝑗(𝐱′) (𝑗 = 𝑅, 𝑍) at

position 𝐱′ = [𝑅 𝑍]𝑇can be obtained from the following boundary integrals over the

nucleus, substratum, and cell membrane boundaries (represented by 𝛤):

𝑐𝑖𝑗𝑣𝑗(𝐱′) + 2π ∫ 𝑣𝑗(𝐱)𝑝𝑖𝑗(𝐱, 𝐱′)𝑅𝑑𝛤(𝐱) = 2π ∫ 𝑇𝑗(𝐱)𝑢𝑖𝑗(𝐱, 𝐱′)𝑅𝑑𝛤(𝐱)𝛤𝛤

4.1

where 𝑢𝑖𝑗(𝐱, 𝐱′) and 𝑝𝑖𝑗(𝐱, 𝐱′) are velocities and tractions, respectively, arising from a

concentrated point force located at position x, given by Kelvin’s fundamental solutions

for the axisymmetric case for linear elasticity (provided in reference [109]), but with the

shear modulus replaced with 𝜇 and the Poisson ratio set to zero. The tensor 𝑐𝑖𝑗 is equal

to the Kronecker delta (identity tensor) 𝛿𝑖𝑗, on the cytosplasmic domain and is a known

tensor on the surface 𝛤. The boundary element method was used to estimate the

Page 106: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

106

instantaneous velocities and stresses at the boundaries [110], such that the evolution of

cell and nuclear shapes could be simulated by numerically integrating the boundary

positions over time. The cell surface was discretized into 100 axisymmetric quadratic

boundary elements and the nuclear surface into 50 elements. Integrals on the elements

were approximated with 10th order Gauss quadrature except strongly singular integrals,

which were obtained from analytical rigid body translation (z-direction) and plan strain

(r-direction) conditions [109]. At each time step, the surface velocities and stresses were

calculated under the constraint of constant volume, and time-stepping was performed

using the 4th-order Runge-Kutta method. The constraint of constant volume was

imposed by simultaneously solving for the hydrostatic pressure 𝑃ℎ at each time step that

keeps the net volume change equal to zero. The node spacing was reset at each time

step by interpolation using cubic Hermite interpolating polynomials (MATLAB function

pchip). To prevent close approach of the nuclear surface to the cell membrane (which

mathematically allowed by way of Eq. A1-1, but creates numerical issues due to surface

singularities in the boundary element method), a close-range repulsive pressure was

imposed for close separation distances 𝑧 of the form 𝑃(𝑧) = (𝑑 𝑧⁄ )3𝑒−𝑧 𝑑⁄ 𝑑 = 0.01 𝑅𝑛.

The initial condition was set to a nearly spherical cell with a small contact area of

approximately 0.5% of the total cell membrane surface area.

Page 107: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

107

Table A-1. Parameters of Cell Spreading Model

Parameter Symbol Value Source

Contractile stress 𝜎𝑐 0.19

nN/m2

Estimated from myosin-induced

nuclear volume change

Nucleus bulk modulus 𝐾 0.25

nN/m2

Ref [11], isolated Xenopus oocyte

nuclei

Nucleus area modulus 𝜅 25 nN/m Ref [11], isolated Xenopus oocyte

nuclei

Membrane tension 𝑇𝑚𝑒𝑚 0.1 nN/m Ref [106], moving fish keratocytes;

Ref [107]

Network Viscosity

Parameter

𝜇 0.21 nN-

s/m2

Ref [108], adherent J774

macrophages

Nuclear lamina bending

stiffness

𝑘𝑐 3.5x10-4

nN-m

Ref [111], MEFs

Network assembly speed

at contact boundary with

substratum

𝑣𝑎𝑐 0.5 m/min Estimated from spreading speed

Network assembly speed

at cell cortex

𝑣𝑎 Varied

Energy parameter in area

expansion equation

𝐸𝑠 3.2x10-4

nN-m

Estimated from excess surface

area

Nuclear radius 𝑅𝑛 6.3 m Measured

Cell radius (rounded) 𝑅 8.3 m Measured

Page 108: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

108

APPENDIX B THE INFLUENCE OF CELL GEOMETRY ON NUCLEAR VOLUME

The evidence that nuclear shape mimics the change of cell shape triggers our

interests on the change of nuclear volume with different cell geometry. Vincent Tocco, a

graduate in my lab, has shown that the nuclear volume decreases significantly in cells

cultured on 1D line comparing with 2D surface, which means that cell geometry is able

to regulate nuclear volume. Then will the nuclear volume increase if the cell geometry is

removed? To answer this question, alive cells with GFP-histone expression on 1D line

and 2D surface were detached from substratum with trypsinization and imaged until

them completely became rounded. For cells on 2D surface, no significant change of

nuclear volume was observed for individual cells along with the decreased cell

spreading area (Figure B-1A). Consistently, average change of nuclear volume does not

change significantly in statistics although there is a tendency of decay (~5% drop)

during the process of detachment (Figure B-1B). However, the nuclear volume of cells

on 1D line increases after detaching from substratum (Figure B-1C) and this increase is

statistically significant (Figure B-1D). Comparing the nuclear volume of cells on 1D line

with 2D surface at different stages of cell detachment, the significant difference of

nuclear volume disappears after removal of the cell geometry (Figure B-1E). Taken

together, the change of nuclear volume induced by cell geometry is reversible without

constrain of the certain cell geometry.

Page 109: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

109

Figure B-1 The difference of nuclear volume induced by cell geometry vanishes after

the removal of geometry constrain. The nuclear volume of individual cell A) remains constant during the cell detachment from 2D substratum accompanying with the decreased spreading area of cell. B) The average nuclear volume (normalized by initial volume before detachment) does not have statistically significant difference among the time points in the whole process. C) The nuclear volume of individual cells continuously increases along with the detachment of cell from 1D line, which is further supported by the increasing average of normalized volume of nucleus in D). E) There is statistically significant difference of nuclear volume between cell on 1D line and 2D surface before and immediately after detachment. Yet, this significant difference continuously decreases until disappear with completely rounded cell. (n=20 and 15 for 2D surface and 1D line, respectively. Data are shown as Mean ± SEM.* indicates p<0.05)

Page 110: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

110

Page 111: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

111

LIST OF REFERENCES

1. Brown, R. (1866). On the Organs and Mode of Fecundation of Orchidex and Asclepiadea. M iscellaneous Botanical Works, 511-514.

2. Mulder, J.W.R., Offerhaus, G.J.A., Defeyter, E.P., Floyd, J.J., Kern, S.E., Vogelstein, B., and Hamilton, S.R. (1992). The Relationship Of Quantitative Nuclear Morphology to Molecular Genetic Alterations in the Adenoma-Carcinoma Sequence of the Large-Bowel. American Journal of Pathology 141, 797-804.

3. Pienta, K.J., and Coffey, D.S. (1991). Correlation of nuclear morphometry with progression of breast-cancer. Cancer 68, 2012-2016.

4. Partin, A.W., Steinberg, G.D., Pitcock, R.V., Wu, L., Piantadosi, S., Coffey, D.S., and Epstein, J.I. (1992). Use of nuclear morphometry, gleason histologic scoring, clinical stage, and age to predict disease-free survival among patients with prostate-cancer. Cancer 70, 161-168.

5. Barateau, A., Vadrot, N., Vicart, P., Ferreiro, A., Mayer, M., Heron, D., Vigouroux, C., and Buendia, B. (2017). A Novel Lamin A Mutant Responsible for Congenital Muscular Dystrophy Causes Distinct Abnormalities of the Cell Nucleus. Plos One 12, 18.

6. Broers, J.L.V., Ramaekers, F.C.S., Bonne, G., Ben Yaou, R., and Hutchison, C.J. (2006). Nuclear lamins: Laminopathies and their role in premature ageing. Physiological Reviews 86, 967-1008.

7. Beale, L.S. (1860). Examination of sputum from a case of cancer of the pharynx and the adjacent parts. ( Arch. Med. Lond.), p. 44.

8. Stuurman, N., Heins, S., and Aebi, U. (1998). Nuclear lamins: Their structure, assembly, and interactions. Journal of Structural Biology 122, 42-66.

9. Gruenbaum, Y., Wilson, K.L., Harel, A., Goldberg, M., and Cohen, M. (2000). Nuclear Lamins - Structural proteins with fundamental functions. Journal of Structural Biology 129, 313-323.

10. Schirmer, E.C., Guan, T.L., and Gerace, L. (2001). Involvement of the lamin rod domain in heterotypic lamin interactions important for nuclear organization. Journal of Cell Biology 153, 479-489.

11. Dahl, K.N., Kahn, S.M., Wilson, K.L., and Discher, D.E. (2004). The nuclear envelope lamina network has elasticity and a compressibility limit suggestive of a molecular shock absorber. Journal of Cell Science 117, 4779-4786.

Page 112: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

112

12. Stierle, V.N., Couprie, J.L., Ostlund, C., Krimm, I., Zinn-Justin, S., Hossenlopp, P., Worman, H.J., Courvalin, J.C., and Duband-Goulet, I. (2003). The carboxyl-terminal region common to lamins A and C contains a DNA binding domain. Biochemistry 42, 4819-4828.

13. Cartwright, S., and Karakesisoglou, I. (2014). Nesprins in health and disease. Seminars in Cell & Developmental Biology 29, 169-179.

14. Matsumoto, A., Hieda, M., Yokoyama, Y., Nishioka, Y., Yoshidome, K., Tsujimoto, M., and Matsuura, N. (2015). Global loss of a nuclear lamina component, lamin A/C, and LINC complex components SUN1, SUN2, and nesprin-2 in breast cancer. Cancer Med.

15. Burke, B., and Stewart, C.L. (2002). Life at the edge: The nuclear envelope and human disease. Nature Reviews Molecular Cell Biology 3, 575-585.

16. Capo-chichi, C.D., Cai, K.Q., Simpkins, F., Ganjei-Azar, P., Godwin, A.K., and Xu, X.X. (2011). Nuclear envelope structural defects cause chromosomal numerical instability and aneuploidy in ovarian cancer. Bmc Medicine 9, 11.

17. Thomas, C.H., Collier, J.H., Sfeir, C.S., and Healy, K.E. (2002). Engineering gene expression and protein synthesis by modulation of nuclear shape. Proceedings of the National Academy of Sciences of the United States of America 99, 1972-1977.

18. Vergani, L., Grattarola, M., and Nicolini, C. (2004). Modifications of chromatin structure and gene expression following induced alterations of cellular shape. International Journal of Biochemistry & Cell Biology 36, 1447-1461.

19. Misteli, T. (2007). Beyond the sequence: Cellular organization of genome function. Cell 128, 787-800.

20. Dahl, K.N., Ribeiro, A.J.S., and Lammerding, J. (2008). Nuclear shape, mechanics, and mechanotransduction. Circulation Research 102, 1307-1318.

21. Fletcher, D.A., and Mullins, D. (2010). Cell mechanics and the cytoskeleton. Nature 463, 485-492.

22. Versaevel, M., Grevesse, T., and Gabriele, S. (2012). Spatial coordination between cell and nuclear shape within micropatterned endothelial cells. Nature Communications 3, 11.

23. Chancellor, T.J., Lee, J., Thodeti, C., and Lele, T.P. (2012). Actomyosin tension exerted on the nucleus through nesprin-1 connections influences endothelial cell adhesion, migration, and cyclic strain induced reorientation (vol 99, pg 115, 2010). Biophysical Journal 102, 2411-2411.

Page 113: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

113

24. Sims, J.R., Karp, S., and Ingber, D.F. (1992). Altering the cellular mechanical force balance results in integrated changes in cell, cytoskeletal and nuclear shape. Journal of Cell Science 103, 1215-1222.

25. Folker, E.S., Ostlund, C., Luxton, G.W.G., Worman, H.J., and Gundersen, G.G. (2011). Lamin A variants that cause striated muscle disease are defective in anchoring transmembrane actin-associated nuclear lines for nuclear movement. Proceedings of the National Academy of Sciences of the United States of America 108, 131-136.

26. Luxton, G.W.G., Gomes, E.R., Folker, E.S., Vintinner, E., and Gundersen, G.G. (2010). Linear Arrays of Nuclear Envelope Proteins Harness Retrograde Actin Flow for Nuclear Movement. Science 329, 956-959.

27. Roux, K.J., Crisp, M.L., Liu, Q., Kim, D., Kozlov, S., Stewart, C.L., and Burke, B. (2009). Nesprin 4 is an outer nuclear membrane protein that can induce kinesin-mediated cell polarization. Proceedings of the National Academy of Sciences of the United States of America 106, 2194-2199.

28. Horn, H.F., Kim, D.I., Wright, G.D., Wong, E.S.M., Stewart, C.L., Burke, B., and Roux, K.J. (2013). A mammalian KASH domain protein coupling meiotic chromosomes to the cytoskeleton. Journal of Cell Biology 202, 1023-1039.

29. Wu, J., Lee, K.C., Dickinson, R.B., and Lele, T.P. (2011). How Dynein and Microtubules Rotate the Nucleus. Journal of Cellular Physiology 226, 2666-2674.

30. Levy, J.R., and Holzbaur, E.L.F. (2008). Dynein drives nuclear rotation during forward progression of motile fibroblasts. Journal of Cell Science 121, 3187-3195.

31. Tremblay, D., Andrzejewski, L., Leclerc, A., and Pelling, A.E. (2013). Actin and Microtubules Play Distinct Roles in Governing the Anisotropic Deformation of Cell Nuclei in Response to Substrate Strain. Cytoskeleton 70, 837-848.

32. Esue, O., Carson, A.A., Tseng, Y., and Wirtz, D. (2006). A direct interaction between actin and vimentin filaments mediated by the tail domain of vimentin. Journal of Biological Chemistry 281, 30393-30399.

33. Dupin, I., Sakamoto, Y., and Etienne-Manneville, S. (2011). Cytoplasmic intermediate filaments mediate actin-driven positioning of the nucleus. Journal of Cell Science 124, 865-872.

34. Tapley, E.C., and Starr, D.A. (2013). Connecting the nucleus to the cytoskeleton by SUN-KASH bridges across the nuclear envelope. Current Opinion in Cell Biology 25, 57-62.

Page 114: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

114

35. Lombardi, M.L., Jaalouk, D.E., Shanahan, C.M., Burke, B., Roux, K.J., and Lammerding, J. (2011). The Interaction between Nesprins and Sun Proteins at the Nuclear Envelope Is Critical for Force Transmission between the Nucleus and Cytoskeleton. Journal of Biological Chemistry 286, 26743-26753.

36. Ostlund, C., Folker, E.S., Choi, J.C., Gomes, E.R., Gundersen, G.G., and Worman, H.J. (2009). Dynamics and molecular interactions of linker of nucleoskeleton and cytoskeleton (LINC) complex proteins. Journal of Cell Science 122, 4099-4108.

37. Alam, S.G., Lovett, D., Kim, D.I., Roux, K.J., Dickinson, R.B., and Lele, T.P. (2015). The nucleus is an intracellular propagator of tensile forces in NIH 3T3 fibroblasts. Journal of Cell Science 128, 1901-1911.

38. Khatau, S.B., Hale, C.M., Stewart-Hutchinson, P.J., Patel, M.S., Stewart, C.L., Searson, P.C., Hodzic, D., and Wirtz, D. (2009). A perinuclear actin cap regulates nuclear shape. Proceedings of the National Academy of Sciences of the United States of America 106, 19017-19022.

39. Larrieu, D., Britton, S., Demir, M., Rodriguez, R., and Jackson, S.P. (2014). Chemical Inhibition of NAT10 Corrects Defects of Laminopathic Cells. Science 344, 527-532.

40. Chow, K.H., Factor, R.E., and Ullman, K.S. (2012). The nuclear envelope environment and its cancer connections. Nature Reviews Cancer 12, 196-209.

41. Dey, P. (2010). Cancer Nucleus: Morphology and Beyond. Diagnostic Cytopathology 38, 382-390.

42. Jevtic, P., and Levy, D.L. (2014). Mechanisms of nuclear size regulation in model systems and cancer. Advances in experimental medicine and biology 773, 537-569.

43. de las Heras, J.I., Batrakou, D.G., and Schirmer, E.C. (2013). Cancer biology and the nuclear envelope: A convoluted relationship. Seminars in Cancer Biology 23, 125-137.

44. True, L.D., and Jordan, C.D. (2008). The cancer nuclear microenvironment: Interface between light microscopic cytology and molecular phenotype. Journal of Cellular Biochemistry 104, 1994-2003.

45. Isermann, P., and Lammerding, J. (2013). Nuclear Mechanics and Mechanotransduction in Health and Disease. Current Biology 23, R1113-R1121.

46. Worman, H.J., and Bonne, G. (2007). "Laminopathies": A wide spectrum of human diseases. Experimental Cell Research 313, 2121-2133.

Page 115: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

115

47. Crisp, M., Liu, Q., Roux, K., Rattner, J.B., Shanahan, C., Burke, B., Stahl, P.D., and Hodzic, D. (2006). Coupling of the nucleus and cytoplasm: role of the LINC complex. Journal of Cell Biology 172, 41-53.

48. Sosa, B.A., Rothballer, A., Kutay, U., and Schwartz, T.U. (2012). LINC Complexes Form by Binding of Three KASH Peptides to Domain Interfaces of Trimeric SUN Proteins. Cell 149, 1035-1047.

49. Gerashchenko, M.V., Chernoivanenko, I.S., Moldaver, M.V., and Minin, A.A. (2009). Dynein is a motor for nuclear rotation while vimentin IFs is a "brake". Cell Biology International 33, 1057-1064.

50. Kuypers, L.C., Decraemer, W.F., Dirckx, J.J.J., and Timmermans, J.P. (2005). A procedure to determine the correct thickness of an object with confocal microscopy in case of refractive index mismatch. Journal of Microscopy-Oxford 218, 68-78.

51. Jurado, C., Haserick, J.R., and Lee, J. (2005). Slipping or gripping? Fluorescent speckle microscopy in fish keratocytes reveals two different mechanisms for generating a retrograde flow of actin. Molecular Biology of the Cell 16, 507-518.

52. Alt, W., and Dembo, M. (1999). Cytoplasm dynamics and cell motion: two-phase flow models. Mathematical Biosciences 156, 207-228.

53. Dembo, M. (1989). MECHANICS AND CONTROL OF THE CYTOSKELETON IN AMEBA-PROTEUS. Biophysical Journal 55, 1053-1080.

54. Dembo, M., and Harlow, F. (1986). Cell Motion, Contractile Networks, and the Physics of Interpenetrating Reactive Flow. Biophysical Journal 50, 109-121.

55. Swift, J., Ivanovska, I.L., Buxboim, A., Harada, T., Dingal, P., Pinter, J., Pajerowski, J.D., Spinler, K.R., Shin, J.W., Tewari, M., et al. (2013). Nuclear Lamin-A Scales with Tissue Stiffness and Enhances Matrix-Directed Differentiation. Science 341, 975-+.

56. Mitchison, T., and Kirschner, M. (1988). Cytoskeletal Dynamics and Nerve Growth. Neuron 1, 761-772.

57. Wu, J., Kent, I.A., Shekhar, N., Chancellor, T.J., Mendonca, A., Dickinson, R.B., and Lele, T.P. (2014). Actomyosin Pulls to Advance the Nucleus in a Migrating Tissue Cell. Biophysical Journal 106, 7-15.

58. Petrie, R.J., Koo, H., and Yamada, K.M. (2014). Generation of compartmentalized pressure by a nuclear piston governs cell motility in a 3D matrix. Science 345, 1062-1065.

Page 116: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

116

59. Gundersen, G.G., and Worman, H.J. (2013). Nuclear Positioning. Cell 152, 1376-1389.

60. Vishavkarma, R., Raghavan, S., Kuyyamudi, C., Majumder, A., Dhawan, J., and Pullarkat, P.A. (2014). Role of actin filaments in correlating nuclear shape and cell spreading. PLoS One 9, e107895.

61. Versaevel, M., Braquenier, J.B., Riaz, M., Grevesse, T., Lantoine, J., and Gabriele, S. (2014). Super-resolution microscopy reveals LINC complex recruitment at nuclear indentation sites. Sci Rep 4, 7362.

62. Lovett, D.B., Shekhar, N., Nickerson, J.A., Roux, K.J., and Lele, T.P. (2013). Modulation of Nuclear Shape by Substrate Rigidity. Cellular and Molecular Bioengineering 6, 230-238.

63. Bussolati, G., Marchio, C., Gaetano, L., Lupo, R., and Sapino, A. (2008). Pleomorphism of the nuclear envelope in breast cancer: a new approach to an old problem. Journal of Cellular and Molecular Medicine 12, 209-218.

64. Wrzolek, M., Kozarski, T., Alastra, A., Mastrangelo, E., and Rosenblum, M. (2009). Papillary Tumor of the Pineal Region With Marked Nuclear Pleomorphism, Non-Papillary Pattern, and Focal Oncocytic Features. Journal of Neuropathology and Experimental Neurology 68, 585-586.

65. Sabo, E., Gibrat, M., Sova, Y., Stein, A., and Resnick, M.B. (2003). Validation of the novel indices of nuclear pleomorphism, polarity and spatial distribution in the grading of urothelial carcinoma. Analytical and Quantitative Cytology and Histology 25, 53-62.

66. Wolberg, W.H., Street, W.N., and Mangasarian, O.L. (1999). Importance of nuclear morphology in breast cancer prognosis. Clinical Cancer Research 5, 3542-3548.

67. Abdalla, F., Boder, J., Markus, R., Hashmi, H., Buhmeida, A., and Collan, Y. (2009). Correlation of Nuclear Morphometry of Breast Cancer in Histological Sections with Clinicopathological Features and Prognosis. Anticancer Research 29, 1771-1776.

68. Dahl, K.N., and Kalinowski, A. (2011). Nucleoskeleton mechanics at a glance. Journal of Cell Science 124, 675-678.

69. Wilson, K.L., and Berk, J.M. (2010). The nuclear envelope at a glance. Journal of Cell Science 123, 1973-1978.

Page 117: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

117

70. He, S.H., Dunn, K.L., Espino, P.S., Drobic, B., Li, L., Yu, J., Sun, J.M., Chen, H.Y., Pritchard, S., and Davie, J.R. (2008). Chromatin organization and nuclear microenvironments in cancer cells. Journal of Cellular Biochemistry 104, 2004-2015.

71. Solovei, I., Wang, A.S., Thanisch, K., Schmidt, C.S., Krebs, S., Zwerger, M., Cohen, T.V., Devys, D., Foisner, R., Peichl, L., et al. (2013). LBR and Lamin A/C Sequentially Tether Peripheral Heterochromatin and Inversely Regulate Differentiation. Cell 152, 584-598.

72. Alam, S.G., Zhang, Q., Prasad, N., Li, Y., Chamala, S., Kuchibhotla, R., Birendra, K.C., Aggarwal, V., Shrestha, S., Jones, A.L., et al. (2016). The mammalian LINC complex regulates genome transcriptional responses to substrate rigidity. Scientific Reports 6, 11.

73. Booth-Gauthier, E.A., Du, V., Ghibaudo, M., Rape, A.D., Dahl, K.N., and Ladoux, B. (2013). Hutchinson-Gilford progeria syndrome alters nuclear shape and reduces cell motility in three dimensional model substrates. Integrative Biology 5, 569-577.

74. Gisselsson, D., Bjork, J., Hoglund, M., Mertens, F., Dal Cin, P., Akerman, M., and Mandahl, N. (2001). Abnormal nuclear shape in solid tumors reflects mitotic instability. American Journal of Pathology 158, 199-206.

75. Zhu, Q., Zheng, F., Liu, A.P., Qian, J., Fu, C.H., and Lin, Y. (2016). Shape Transformation of the Nuclear Envelope during Closed Mitosis. Biophysical Journal 111, 2309-2316.

76. Potapova, T.A., Zhu, J., and Li, R. (2013). Aneuploidy and chromosomal instability: a vicious cycle driving cellular evolution and cancer genome chaos. Cancer and Metastasis Reviews 32, 377-389.

77. Funkhouser, C.M., Sknepnek, R., Shimi, T., Goldman, A.E., Goldman, R.D., and de la Cruz, M.O. (2013). Mechanical model of blebbing in nuclear lamin meshworks. Proceedings of the National Academy of Sciences of the United States of America 110, 3248-3253.

78. Li, Y., Lovett, D., Zhang, Q., Neelam, S., Kuchibhotla, R.A., Zhu, R.J., Gundersen, G.G., Lele, T.P., and Dickinson, R.B. (2015). Moving Cell Boundaries Drive Nuclear Shaping during Cell Spreading. Biophysical Journal 109, 670-686.

79. Schindelin, J., Arganda-Carreras, I., Frise, E., Kaynig, V., Longair, M., Pietzsch, T., Preibisch, S., Rueden, C., Saalfeld, S., Schmid, B., et al. (2012). Fiji: an open-source platform for biological-image analysis. Nature Methods 9, 676-682.

Page 118: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

118

80. Kim, D.I., Birendra, K.C., and Roux, K.J. (2015). Making the LINC: SUN and KASH protein interactions. Biological Chemistry 396, 295-310.

81. Chang, W., Worman, H.J., and Gundersen, G.G. (2015). Accessorizing and anchoring the LINC complex for multifunctionality. Journal of Cell Biology 208, 11-22.

82. Cerami, E., Gao, J.J., Dogrusoz, U., Gross, B.E., Sumer, S.O., Aksoy, B.A., Jacobsen, A., Byrne, C.J., Heuer, M.L., Larsson, E., et al. (2012). The cBio Cancer Genomics Portal: An Open Platform for Exploring Multidimensional Cancer Genomics Data. Cancer Discovery 2, 401-404.

83. Gao, J.J., Aksoy, B.A., Dogrusoz, U., Dresdner, G., Gross, B., Sumer, S.O., Sun, Y.C., Jacobsen, A., Sinha, R., Larsson, E., et al. (2013). Integrative Analysis of Complex Cancer Genomics and Clinical Profiles Using the cBioPortal. Science Signaling 6, 19.

84. Lammerding, J., Fong, L.G., Ji, J.Y., Reue, K., Stewart, C.L., Young, S.G., and Lee, R.T. (2006). Lamins A and C but not lamin B1 regulate nuclear mechanics. Journal of Biological Chemistry 281, 25768-25780.

85. Imbalzano, K.M., Cohet, N., Wu, Q., Underwood, J.M., Imbalzano, A.N., and Nickerson, J.A. (2013). Nuclear Shape Changes Are Induced by Knockdown of the SWI/SNF ATPase BRG1 and Are Independent of Cytoskeletal Connections. Plos One 8, 14.

86. Verboon, J.M., Rincon-Arano, H., Werwie, T.R., Delrow, J.J., Scalzo, D., Nandakumar, V., Groudine, M., and Parkhurst, S.M. (2015). Wash Interacts with Lamin and Affects Global Nuclear Organization. Current Biology 25, 804-810.

87. Tocco, V., Li, Y., Christopher, K., Matthews, J., Aggarwal, V., Paschall, L., Luesch, H., Licht, J., Dickinson, R., and Lele, T. (2017). The nucleus is irreversibly shaped by motion of cell boundaries in cancer and non-cancer cells. Journal of Cellular Physiology. In press.

88. Cremer, T., and Cremer, C. (2001). Chromosome territories, nuclear architecture and gene regulation in mammalian cells. Nature Reviews Genetics 2, 292-301.

89. Xiao, B.T., Freedman, B.S., Miller, K.E., Heald, R., and Marko, J.F. (2012). Histone H1 compacts DNA under force and during chromatin assembly. Molecular Biology of the Cell 23, 4864-4871.

90. Tajik, A., Zhang, Y.J., Wei, F.X., Sun, J., Jia, Q., Zhou, W.W., Singh, R., Khanna, N., Belmont, A.S., and Wang, N. (2016). Transcription upregulation via force-induced direct stretching of chromatin. Nature Materials 15, 1287-1296.

Page 119: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

119

91. Sansregret, L., and Swanton, C. (2017). The Role of Aneuploidy in Cancer Evolution. Cold Spring Harbor Perspectives in Medicine 7, 17.

92. Rajagopalan, H., and Lengauer, C. (2004). Aneuploidy and cancer. Nature 432, 338-341.

93. Neelam, S., Hayes, P.R., Zhang, Q., Dickinson, R.B., and Lele, T.P. (2016). Vertical uniformity of cells and nuclei in epithelial monolayers. Scientific Reports 6, 10.

94. Denais, C.M., Gilbert, R.M., Isermann, P., McGregor, A.L., te Lindert, M., Weigelin, B., Davidson, P.M., Friedl, P., Wolf, K., and Lammerding, J. (2016). Nuclear envelope rupture and repair during cancer cell migration. Science 352, 353-358.

95. Friedl, P., Wolf, K., and Lammerding, J. (2011). Nuclear mechanics during cell migration. Current Opinion in Cell Biology 23, 55-64.

96. Stephens, A.D., Banigan, E.J., Adam, S.A., Goldman, R.D., and Marko, J.F. (2017). Chromatin and lamin A determine two different mechanical response regimes of the cell nucleus. Molecular biology of the cell.

97. Davidson, P.M., Sliz, J., Isermann, P., Denais, C., and Lammerding, J. (2015). Design of a microfluidic device to quantify dynamic intra-nuclear deformation during cell migration through confining environments. Integrative Biology 7, 1534-1546.

98. Allena, R., Thiam, H., Piel, M., and Aubry, D. (2015). A mechanical model to investigate the role of the nucleus during confined cell migration. Computer Methods in Biomechanics and Biomedical Engineering 18, 1868-1869.

99. Jean, R.P., Gray, D.S., Spector, A.A., and Chen, C.S. (2004). Characterization of the nuclear deformation caused by changes in endothelial cell shape. Journal of Biomechanical Engineering-Transactions of the Asme 126, 552-558.

100. Kim, D.H., Li, B., Si, F.W., Phillip, J.M., Wirtz, D., and Sun, S.X. (2015). Volume regulation and shape bifurcation in the cell nucleus. Journal of Cell Science 128, 3375-3385.

101. Li, Q.S., Kumar, A., Makhija, E., and Shivashankar, G.V. (2014). The regulation of dynamic mechanical coupling between actin cytoskeleton and nucleus by matrix geometry. Biomaterials 35, 961-969.

102. Hatch, E.M., and Hetzer, M.W. (2016). Nuclear envelope rupture is induced by actin-based nucleus confinement. Journal of Cell Biology 215, 27-36.

Page 120: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

120

103. Neelam, S., Chancellor, T.J., Li, Y., Nickerson, J.A., Roux, K.J., Dickinson, R.B., and Lele, T.P. (2015). Direct force probe reveals the mechanics of nuclear homeostasis in the mammalian cell. Proceedings of the National Academy of Sciences of the United States of America 112, 5720-5725.

104. Evans, E., and Rawicz, W. (1990). Entropy-Driven Tension and Bending Elasticity in Condensed-Fluid Membranes. Physical Review Letters 64, 2094-2097.

105. Brangwynne, C.P., MacKintosh, F.C., and Weitz, D.A. (2007). Force fluctuations and polymerization dynamics of intracellular microtubules. Proceedings of the National Academy of Sciences of the United States of America 104, 16128-16133.

106. Lieber, A.D., Yehudai-Resheff, S., Barnhart, E.L., Theriot, J.A., and Keren, K. (2013). Membrane Tension in Rapidly Moving Cells Is Determined by Cytoskeletal Forces. Current Biology 23, 1409-1417.

107. Gauthier, N.C., Masters, T.A., and Sheetz, M.P. (2012). Mechanical feedback between membrane tension and dynamics. Trends in Cell Biology 22, 527-535.

108. Bausch, A.R., Moller, W., and Sackmann, E. (1999). Measurement of local viscoelasticity and forces in living cells by magnetic tweezers. Biophysical Journal 76, 573-579.

109. Bakr, A.A. (1985). The Boundary Integral Equation Method in Axisymmetric Stress Analysis Problems Volume 14 (Springer-Verlag).

110. Brebbia, C.A., Telles, J.C.F., and Wrobel, L.C. (1984). Boundary Element Techniques. In Theory and Applications in Engineering. (Springer Berlin Heidelberg).

111. Vaziri, A., Lee, H., and Mofrad, M.R.K. (2006). Deformation of the cell nucleus under indentation: Mechanics and mechanisms. Journal of Materials Research 21, 2126-2135.

Page 121: By YUAN LI - ufdcimages.uflib.ufl.eduufdcimages.uflib.ufl.edu/UF/E0/05/13/82/00001/LI_Y.pdf · David Lovett who trained me on all basic techniques in the lab and helped me design

121

BIOGRAPHICAL SKETCH

Yuan Li was born in Baoji City, Shaanxi Province, China in 1988 to Yinchi Liu

and Heng Li. He finished his high school from Baoji Middle School, China in 2006. In

July of 2010, he received the Bachelor of Science degree in chemistry from College of

Science, Nanchang University, Jiangxi, China. He joined University of Florida in 2011

and conducted research on mechanics of cell and nucleus in Dr. Tanmay Lele’s lab in

the Department of Chemical Engineering. He earned his Master of Science degree in

chemical engineering in 2013. In 2014, he converted to PhD program in Dr. Tanmay

Lele’s group and continued his research on studying mechanisms of nuclear shaping in

fibroblasts, epithelial cells and breast cancer cells. He earned his Doctor of Philosophy

in chemical engineering from the University of Florida in 2017.


Recommended