+ All Categories
Home > Documents > Developmental Cell Perspective - COnnecting REpositories · Developmental Cell Perspective Tracing...

Developmental Cell Perspective - COnnecting REpositories · Developmental Cell Perspective Tracing...

Date post: 26-May-2020
Category:
Upload: others
View: 4 times
Download: 0 times
Share this document with a friend
16
Developmental Cell Perspective Tracing Cells for Tracking Cell Lineage and Clonal Behavior Margaret E. Buckingham 1, * and Sigole ` ne M. Meilhac 1, * 1 Molecular Genetics of Development Unit, CNRS URA 2578, Department of Developmental Biology, Institut Pasteur, 28 rue du Dr Roux, 75015 Paris, France *Correspondence: [email protected] (M.E.B.), [email protected] (S.M.M.) DOI 10.1016/j.devcel.2011.07.019 Reconstructing the lineage of cells is central to understanding development and is now also an important issue in stem cell research. Technological advances in genetically engineered permanent cell labeling, together with a multiplicity of fluorescent markers and sophisticated imaging, open new possibilities for prospective and retrospective clonal analysis. History and Concepts Cell lineage analysis is intimately connected with the emergence of developmental biology as a field of scientific research (see Galperin, 1998). In the mid-19th century, identification of cells as the basic units of life (Schleiden, 1838; Schwann, 1839) led to the realization that cells come from pre-existing cells (Virchow, 1858). Before the end of the 19th century, the work of Charles Whitman (1887) and later of Edmund Wilson (1892) on leech and annelid embryos led to the formulation of the term ‘‘cell lineage.’’ This early work inspired the Wood’s Hole School at the Marine Biology Laboratory in Massachusetts, where pioneer- ing research in invertebrate embryos led to important concepts for lineage analysis. Thus E. Wilson viewed lineage in terms of the fate of cells and E.G. Conklin (1905), another major figure, made the distinction between determinate and indeterminate types of cleavage in ascidians, leading to the concept of invariant and noninvariant cell lineages. Breakthroughs in vertebrate fate mapping came from the systematic use of vital staining of groups of cells (Vogt, 1929) and from grafting experiments (Spemann and Mangold, 1924) in the amphibian embryo. In addition to embryological approaches, the work of A.H. Sturtevant, based on genetic studies initiated by T.H. Morgan and others on spon- taneously generated mosaicism in insects, led to retrospective analyses in which cell lineage and gene function were associated (Sturtevant, 1929). Many of the conceptual issues of today were evident when cell lineages were first explored. Lineage studies, then as now, aim to establish which cells, and how many cells, in the early embryo will give rise to a structure and, as development proceeds, from which part of a structure a substructure derives. These interrogations now extend to the origin of stem cells that permit the regeneration of an adult structure as well as its initial forma- tion. Clonal analyses, which describe the derivatives of a single cell, provide insight into the mode of growth of a tissue and its regionalization with potential clonal boundaries (Garcia-Bellido et al., 1973) between compartments, or with segregation between distinct cell lineages, which do not necessarily corre- spond to distinct differentiated cell types but rather to topo- graphical subdivisions (Lescroart et al., 2010). Analyses of clones can also provide information about cell death and prolif- eration, cell competition, cell movement and dispersion, and tissue polarity. Experimentation in a growing number of tissues and model organisms reveals the diversity of cell behavior that underlies progression along a lineage tree and has led to the elaboration of conceptual frameworks for cell lineage analysis (e.g., Garcia-Bellido, 1985; Petit et al., 2005; Stent, 1985). In the context of embryonic development, many invertebrates have invariant lineages, meaning that a blastomere not only has a predictable future but also has a reproducible position and a defined group of neighbors from one individual to another. This is illustrated by C. elegans, for which a complete lineage tree has been defined (Sulston et al., 1983). In contrast, in the early mouse embryo, for example, more cell mixing takes place and cells in the inner cell mass of the blastocyst retain pluripo- tency and plasticity (Cockburn and Rossant, 2010). In the case of such noninvariant (regulative) development it is more chal- lenging to analyze cell lineages. Intertwined with the concept of lineage is that of cell commit- ment. Cell lineage follows the normal fate of a cell and its daugh- ters, leading to the formulation of genealogical trees of cells with increasingly restricted cell fate choices as development proceeds. Unlike lineage, commitment can only be established by experimental challenge, such as ectopic grafting or in vitro manipulation, showing that the cell has acquired a restricted cell fate potential (e.g., Tam et al., 1997). As G. Stent (1985) pointed out, cell lineage plays a role in cell commitment by the unequal partitioning of cell determinants in daughter cells in successive cell divisions, as illustrated by the ascidian, Ciona (Nishida, 1987), and by the orderly placement of cells relative to intercellular signals as development proceeds, which is a major feature of vertebrate embryogenesis. It has become increasingly clear that even differentiated cells retain plasticity, as demonstrated by the spectacular phenomenon of induced pluripotency (Yamanaka, 2009). Caution should be exercised in equating cell fate restriction with gene expression. Character- izing lineage progression in these terms provides a genetic complement to cellular studies but can also lead to experimental pitfalls as discussed later. Experimentalists today face many of the same dilemmas that confronted embryologists a hundred years ago—namely, how to label cells and subsequently analyze their contribution to the embryo based on the perdurance of the label, without perturbing the development of the organism. New technological develop- ments now facilitate detailed analysis of complex situations. 394 Developmental Cell 21, September 13, 2011 ª2011 Elsevier Inc.
Transcript
Page 1: Developmental Cell Perspective - COnnecting REpositories · Developmental Cell Perspective Tracing Cells for Tracking Cell Lineage and Clonal Behavior Margaret E. Buckingham1, *and

Developmental Cell

Perspective

Tracing Cells for TrackingCell Lineage and Clonal Behavior

Margaret E. Buckingham1,* and Sigolene M. Meilhac1,*1Molecular Genetics of Development Unit, CNRS URA 2578, Department of Developmental Biology, Institut Pasteur, 28 rue du Dr Roux,75015 Paris, France*Correspondence: [email protected] (M.E.B.), [email protected] (S.M.M.)DOI 10.1016/j.devcel.2011.07.019

Reconstructing the lineage of cells is central to understanding development and is now also an importantissue in stem cell research. Technological advances in genetically engineered permanent cell labeling,together with a multiplicity of fluorescent markers and sophisticated imaging, open new possibilities forprospective and retrospective clonal analysis.

History and ConceptsCell lineage analysis is intimately connected with the emergence

of developmental biology as a field of scientific research (see

Galperin, 1998). In the mid-19th century, identification of cells

as the basic units of life (Schleiden, 1838; Schwann, 1839) led

to the realization that cells come frompre-existing cells (Virchow,

1858). Before the end of the 19th century, the work of Charles

Whitman (1887) and later of Edmund Wilson (1892) on leech

and annelid embryos led to the formulation of the term ‘‘cell

lineage.’’ This early work inspired the Wood’s Hole School at

theMarine Biology Laboratory inMassachusetts, where pioneer-

ing research in invertebrate embryos led to important concepts

for lineage analysis. Thus E. Wilson viewed lineage in terms of

the fate of cells and E.G. Conklin (1905), another major figure,

made the distinction between determinate and indeterminate

types of cleavage in ascidians, leading to the concept of invariant

and noninvariant cell lineages. Breakthroughs in vertebrate fate

mapping came from the systematic use of vital staining of groups

of cells (Vogt, 1929) and from grafting experiments (Spemann

and Mangold, 1924) in the amphibian embryo. In addition to

embryological approaches, the work of A.H. Sturtevant, based

on genetic studies initiated by T.H. Morgan and others on spon-

taneously generated mosaicism in insects, led to retrospective

analyses in which cell lineage and gene function were associated

(Sturtevant, 1929).

Many of the conceptual issues of today were evident when cell

lineageswere first explored. Lineage studies, then as now, aim to

establish which cells, and how many cells, in the early embryo

will give rise to a structure and, as development proceeds,

from which part of a structure a substructure derives. These

interrogations now extend to the origin of stem cells that permit

the regeneration of an adult structure as well as its initial forma-

tion. Clonal analyses, which describe the derivatives of a single

cell, provide insight into the mode of growth of a tissue and its

regionalization with potential clonal boundaries (Garcia-Bellido

et al., 1973) between compartments, or with segregation

between distinct cell lineages, which do not necessarily corre-

spond to distinct differentiated cell types but rather to topo-

graphical subdivisions (Lescroart et al., 2010). Analyses of

clones can also provide information about cell death and prolif-

eration, cell competition, cell movement and dispersion, and

tissue polarity. Experimentation in a growing number of tissues

394 Developmental Cell 21, September 13, 2011 ª2011 Elsevier Inc.

and model organisms reveals the diversity of cell behavior that

underlies progression along a lineage tree and has led to the

elaboration of conceptual frameworks for cell lineage analysis

(e.g., Garcia-Bellido, 1985; Petit et al., 2005; Stent, 1985).

In the context of embryonic development, many invertebrates

have invariant lineages, meaning that a blastomere not only has

a predictable future but also has a reproducible position and

a defined group of neighbors from one individual to another.

This is illustrated by C. elegans, for which a complete lineage

tree has been defined (Sulston et al., 1983). In contrast, in the

early mouse embryo, for example, more cell mixing takes place

and cells in the inner cell mass of the blastocyst retain pluripo-

tency and plasticity (Cockburn and Rossant, 2010). In the case

of such noninvariant (regulative) development it is more chal-

lenging to analyze cell lineages.

Intertwined with the concept of lineage is that of cell commit-

ment. Cell lineage follows the normal fate of a cell and its daugh-

ters, leading to the formulation of genealogical trees of cells

with increasingly restricted cell fate choices as development

proceeds. Unlike lineage, commitment can only be established

by experimental challenge, such as ectopic grafting or in vitro

manipulation, showing that the cell has acquired a restricted

cell fate potential (e.g., Tam et al., 1997). As G. Stent (1985)

pointed out, cell lineage plays a role in cell commitment by the

unequal partitioning of cell determinants in daughter cells in

successive cell divisions, as illustrated by the ascidian, Ciona

(Nishida, 1987), and by the orderly placement of cells relative

to intercellular signals as development proceeds, which is

a major feature of vertebrate embryogenesis. It has become

increasingly clear that even differentiated cells retain plasticity,

as demonstrated by the spectacular phenomenon of induced

pluripotency (Yamanaka, 2009). Caution should be exercised in

equating cell fate restriction with gene expression. Character-

izing lineage progression in these terms provides a genetic

complement to cellular studies but can also lead to experimental

pitfalls as discussed later.

Experimentalists today face many of the same dilemmas that

confronted embryologists a hundred years ago—namely, how to

label cells and subsequently analyze their contribution to the

embryo based on the perdurance of the label, without perturbing

the development of the organism. New technological develop-

ments now facilitate detailed analysis of complex situations.

Page 2: Developmental Cell Perspective - COnnecting REpositories · Developmental Cell Perspective Tracing Cells for Tracking Cell Lineage and Clonal Behavior Margaret E. Buckingham1, *and

Developmental Cell

Perspective

In the following sections we shall discuss the current state of

this art and future developments, where temporal as well as

spatial regulation of the onset of labeling, simultaneous detec-

tion of several lineages, systematic labeling of all progenitors

of a structure, visualization of the dynamics of lineage progres-

sion, and linking lineage to gene function are the underlying

issues. In this era of genetic tools for cell tracing, we will focus

on Drosophila, zebrafish, and mouse, with reference to avians,

amphibians, and plants, as well as to the other invertebrate

models that have provided important insights into lineage anal-

ysis. Approaches currently employed for following the history

of a cell, which we discuss here, are summarized in Table 1.

Prospective Lineage Analysis: Selectionof Labeled ProgenitorsProspective lineage analysis is a classic approach in which cell

labeling is performed at a known position and stage and the

contribution of the cell’s descendants to a structure is subse-

quently analyzed. In this context, cell fate mapping can be

achieved by grafting experiments where labeled cells are intro-

duced into the embryo and their subsequent contributions moni-

tored. Following on from the pioneering work of Mangold and

Spemann in amphibians (Spemann and Mangold, 1924) and of

Waddington in the chick (Waddington, 1932), the analysis of

chick-quail chimaeras, in which it is possible to distinguish nuclei

between these closely related species, underlies important

aspects of our understanding of cell fates in the amniote embryo,

as exemplified by neural crest cell derivatives (Le Douarin and

Barq, 1969). In an alternative, generally applicable approach,

radioactive labeling of transplanted cells has been used

(Weston, 1963) and was instrumental in mapping the heart-form-

ing fields in the chick embryo (Rosenquist and De Haan, 1966).

This approach was also used in challenging experiments on

the mouse embryo to map the fate of cells in the epiblast

(Beddington, 1981). Lypophilic carbocyanine dyes, such as DiI,

which can be introduced into a small region of the embryo

have been, and continue to be, used to trace groups of cells

and examine cell movements. Less invasive than grafting exper-

iments, these vital dyes intercalate into the cell membrane

and are easily visualized. In early experiments with this method

of cell marking, the migration pathways of neural crest cells

and the temporal order in which they contribute to their deriva-

tives were refined for the chick embryo (Serbedzija et al.,

1989). Numerous fate maps have been established using this

technique, including more recently in the lamprey (McCauley

and Bronner-Fraser, 2003) and in cultured mouse embryos (Galli

et al., 2008).

Single-cell labeling, which permits lineage analysis, is more

challenging but can be achieved by microinjection. Classically,

horseradish peroxidase (HRP) or dextran linked fluorescent

dyes, which are too large to diffuse between cells, have been

used as intracellular markers. Pioneering experiments using

HRP pressure mediated microinjection in the leech embryo,

where there is little cell migration (Weisblat et al., 1978), showed

that teloblasts, which are the founder cells of segments, give rise

to topographically invariant lineages that consist of different cell

types. Interestingly, in this case, unlike that of Drosophila (Gar-

cia-Bellido, 1985), morphological segment boundaries do not

necessarily correspond to borders of clonal restriction. In the

De

frog embryo, the progeny of different blastomeres were shown

to contribute to distinct clonal domains with well-defined bound-

aries in the central nervous system (Hirose and Jacobson, 1979),

although the descendants of a blastomere are not restricted to

a single neural, or other, cell fate, indicative of global cell mixing

(Moody, 1987). As distinct from amphibians, early planes of

cleavage are not related to the plane of bilateral symmetry of

the zebrafish embryo, and descendants of a single blastomere

tend to remain associated initially (Kimmel and Law, 1985a) until

they disperse at the onset of epiboly (Kimmel and Law, 1985b).

During gastrulation, cell mixing decreases and tissue-specific

lineages have been observed from this stage (Kimmel and

Warga, 1986). In the mouse embryo, where there is extensive

cell mixing, single-cell labeling by iontophoresis microinjection

of HRP (pioneered by Ba1akier and Pedersen, 1982) in cells of

the epiblast has led to fate maps in which the probability of

descendants of a cell contributing to a particular tissue was

determined (Lawson et al., 1991). In the absence of stereotyped

lineages and despite geometrical differences, topological fate

relationships at the stage of gastrulation are conserved between

mammals, birds, amphibians, and zebrafish.

The advent of fluorescent proteins as markers (Shimomura

et al., 1962) has had a major impact on fate mapping and cell

tracking, as they are genetically encoded. Furthermore, fusion

proteins, inwhich thefluorescentprotein is targeted to thenucleus

(e.g., H2B-GFP) or to the plasma membrane, provide clearer

cellular resolution and additional information, such as mitotic

status or cell shape dynamics. Thus, for example, microinjection

of DNA encoding a fluorescent protein, GFP, demonstrated that

a single cell in the chick somite is bipotent and revealed how its

descendants progressively acquire a dermal or muscle cell fate

(Ben-Yair and Kalcheim, 2005). In another example, microinjec-

tion of mRNAs encoding membrane-bound fluorescent proteins

into a single cell of the inner cell mass of the mouse blastocyst,

followed by time-lapse imaging in relation to a chromosomal

marker, has shown how segregation between epiblast and

primitive endoderm lineages is accompanied by extensive cell

movement and, coupled with early markers of these cell types,

supports the conclusion that primitive endoderm formation

involves cell sorting and position-dependant induction (Meilhac

et al., 2009). In this case, the characteristics of clones were

used to test computer models of mechanisms for lineage segre-

gation. In invertebratemodels, too, fluorescent proteins are being

used to track cells. In the leech embryo, an analysis based on

injection of a plasmid encoding H2B-GFP now indicates a transi-

tion from tightly regulated tomorestochastic cell division, pointing

to a less black-and-white distinction between invariant andnonin-

variant lineages (Gline et al., 2009), as observed to some extent

even in C. elegans (Schnabel et al., 1997).

Advances in understanding chromophore photochemistry

have made it possible to engineer photomodulatable fluorescent

proteins (see Piatkevich et al., 2010), which have tended to

replace caged molecules that require chemical synthesis, for

marker activation. The value of such a caged dyewas first shown

in an experiment in Drosophila that revealed clonal restriction

anteriorly but not posteriorly when the dye was activated at the

site of establishment of an Engrailed 1-positive parasegment

(Vincent and O’Farrell, 1992). In the last few years a range of

photoconvertible fluorescent proteins, which undergo a spectral

velopmental Cell 21, September 13, 2011 ª2011 Elsevier Inc. 395

Page 3: Developmental Cell Perspective - COnnecting REpositories · Developmental Cell Perspective Tracing Cells for Tracking Cell Lineage and Clonal Behavior Margaret E. Buckingham1, *and

Table 1. An Assessment of Currently Used Cell-Tracing Techniques

Organism

Current Methods Requirements Merits Limitations M C F Z D I P

Prospective

DiI vital staining, embryo

or explant culture

targeted, easier than

microinjection

nonclonal, dilution,

accessibility to cells

x x x x

HRP, dextran

microinjection

micromanipulation,

embryo or explant culture

clonal, targeted dilution, invasive,

accessibility to cells

x x x x x

DNA, RNA

microinjection

micromanipulation,

embryo or explant culture

clonal, targeted,

amplification of

the marker

dilution, invasive,

accessibility to cells

x x x x x

Uncaging or

photomodulation of

a fluorescent protein

laser fluorescent

microscopy, embryo or

explant culture, injection

of the marker or

genetically modified line

targeted, no

micromanipulation

dilution, phototoxicity,

accessibility to cells

x x x x

Genetic tracing /

tissue-specific

recombinase

genetically modified lines noninvasive,

permanent

nonclonal, dependent

on gene/promoter

expression and potential

integration site effects

x x x

Transplantations micromanipulation,

labeled donor

permanent if genetic

marker, easier than

microinjection

nonclonal, invasive,

accessibility to cells

x x x x

Mosaic

Early chimaeras

with cell mixing

micromanipulation, lines

with distinct phenotypes

permanent, sparse

labeling

nonclonal, invasive x

DNA electroporation electroporation, cell

tracking

multicolor, sparse

labeling

nonclonal, dilution,

accessibility

x x

X-inactivation genetically

modified lines

spontaneous,

permanent, sparse

labeling

nonclonal x

Multicolor genetic

mosaics: MADM,

twin-spot,

brainbow/confetti

genetically modified

lines, resolving

color hues

multicolor,

permanent,

sparse labeling

nonclonal x x

Retrospective

Spatially Random Labeling

Retrovirus library of tagged

retroviruses, isolation

of cells for PCR/

sequencing analysis

time-control,

permanent, clonal

differential infectivity

of cells, potential

integration site effects

x x

Inducible

recombinase:

temperature,

hormone or

antibiotics

genetically modified

lines, temperature shift

or inducer molecule

concentration, control

background levels

time-control, dose

control of clone

frequency

toxicity, partial activity

of the Cre,

reproducibility

x x x x

Inducible transposon

mobility: temperature,

inducer molecule

genetically modified

lines, temperature shift

or inducer molecule

concentration, control

background levels

time-control, dose

control of clone

frequency

instability, potential

integration site effects

x

Spatially and Temporally Random Labeling

Microsatellites isolation of cells,

sequencing analysis

systematic,

spontaneous

large number of

observations for

statistical analysis

x

Mitotic recombination

with a laacZ-like

reporter

genetically

modified line

systematic,

spontaneous

large number of

observations for

statistical analysis

x x

396 Developmental Cell 21, September 13, 2011 ª2011 Elsevier Inc.

Developmental Cell

Perspective

Page 4: Developmental Cell Perspective - COnnecting REpositories · Developmental Cell Perspective Tracing Cells for Tracking Cell Lineage and Clonal Behavior Margaret E. Buckingham1, *and

Table 1. Continued

Organism

Current Methods Requirements Merits Limitations M C F Z D I P

4D Imaging

Time-lapse imaging embryo or explant

culture, high-resolution

microscopy,

computing capacity

dynamic, direct,

comprehensive

limited developmental

window, penetration,

complexity of image

analysis

x x x x x x

This table is mainly based on papers discussed in the text. M, mouse; C, chick; F, frog; Z, zebrafish; D, Drosophila; I, other invertebrates; P, plants.

Developmental Cell

Perspective

change after exposure to activating light, has become available.

These proteins can be photoconverted by confocal laser micros-

copy or even using regular fluorescence microscopes (Baker

et al., 2010; Stark and Kulesa, 2007). In some instances, photo-

conversion by two-photon microscopy may be applicable (Hatta

et al., 2006) for single-cell labeling, with the advantage of deeper

penetration into the tissue and less phototoxicity. Issues in using

fluorescent proteins for cell tracking include the rapidity and

stability of photoconversion, the brightness of the fluorescence,

and toxicity; it is important to test these parameters for the

organism and developmental stage under study (e.g., Nowot-

schin and Hadjantonakis, 2009). The zebrafish embryo because

of its accessibility, transparency, and rapid development partic-

ularly lends itself to fluorescent cell tracking. Temporal conver-

sion of fluorescent proteins can be used for in vivo birthdating

of tissue types. This is illustrated by the BAPTI system where

a photoconvertible Kaede reporter is under the control of a neural

promoter; after exposure to activating light early-born neurons

B B’

A

early late

Green State Red Stat

Intact polypeptide Backbone cl

Photoconversion

Figure 1. Live Imaging of Kaede Photoconversion in Zebrafish: An Exa(A) Photoconversion of the Kaede fluorescent protein by exposure to UV lighzeiss-campus.magnet.fsu.edu/articles/probes/highlighterfps.html).(B) Early trigeminal sensory neurons born before the photoconversion appear ysynthesized protein (green), whereas the neurons born later remain green.(C) Schematic representation of the mode of action of this BAPTI system on cell

De

are labeled red, whereas later-born neurons, not exposed to

photoconversion, remain green (Figure 1). The BAPTISM system

extends this to include an additional population-specific

reporter. This analysis led to the conclusion that the specification

and function of different classes of trigeminal sensory ganglia

depend on the timing of neurogenesis (Caron et al., 2008).

With the approaches of prospective clonal analysis, fate maps

can be drawn and partial lineages reconstructed. However, the

use of microinjected markers or sequences encoding photomo-

dulatable fluorescent proteins is limited by the problem ofmarker

dilution at each cell division and thus is only applicable for

short-term labeling experiments. Furthermore, their introduction

is invasive, is often challenging technically, and may cause

damage. A major interest of fluorescent proteins is that they

can be employed after stable integration of their coding

sequence into the genome by transgenesis or gene targeting

and can therefore provide permanent cell labeling. This is

classical in mouse and fly and is now becoming practicable in

B’’

405nm

early-born late-born

C e

eaved

mple of Prospective Lineage Analysist induces rupture of a covalent bond (adapted with permission from http://

ellow, due to the presence of the photoconverted red protein and the newly

lineage (Caron et al., 2008, adapted with permission).

velopmental Cell 21, September 13, 2011 ª2011 Elsevier Inc. 397

Page 5: Developmental Cell Perspective - COnnecting REpositories · Developmental Cell Perspective Tracing Cells for Tracking Cell Lineage and Clonal Behavior Margaret E. Buckingham1, *and

Cre Neo Gal4VP16 nlsRFP

hsp70

EF1α SAUVMC

2

B

reporterSTOP

ubiquitous

GIFM or G-TRACE

GFP Gal4VP16

UAS

autoactivation

1-autoexcision

Kaloop

MAZe

C

D

tissue-Gal4 and UAS-Flp ortissue-Cre

A

pA pApA

-expression 3-induction

Figure 2. Genetic Manipulations forSpatiotemporal Control of ReporterExpression(A–C) Schematic representation of genetic tracingprocedures based on tissue-specific expression.(A) Schematic representation of themode of actionof such procedures.(B) Genetic inducible fate mapping (GIFM) used inmouse or G-TRACE in Drosophila depends uponactivation of a ubiquitous reporter by a tissue-specific recombinase (Zinyk et al., 1998; Evanset al., 2009). <: target sites for recombination.(C) In the Kaloop system, as used in zebrafish,autoactivation of the fluorescent marker providespermanent labeling after tissue-specific initiationof expression, without recombination (Distel et al.,2009).(D) In theMAZe procedure, developed in zebrafish,transient activation of a heat-shock promoter (hsp)leads to expression of a reporter via the Gal4/UASsystem (Collins et al., 2010). <: loxP sites for Crerecombination.

Developmental Cell

Perspective

zebrafish, based on transposon-mediated integration. Avian

transgenic lines, based on lentiviral-mediated transgene integra-

tion, should also provide new tools for lineage analysis (Sato

et al., 2010). Further improvements for directing transgene

insertion in a range of species can be envisaged with zinc finger

nucleases, meganucleases, or TALE nucleases (Christian et al.,

2010).

Genetic TracingIn order to follow the descendants of a cell, the recombinase

approach to permanent genetic labeling by specific activation

of a conditional reporter is widely used in mouse and fly and is

now available in fish and has also been used for genetic cell

tracing in Xenopus (Satoh et al., 2005). Recombinase activity

should be rapid, efficient, and specific, although there can be

problems with certain loci and with Cre toxicity, even in mice

(Naiche and Papaioannou, 2007). This is a problem in

Drosophila, where Flp recombinases are the preferred tools.

Improved variants of Flp and Cre, together with the identification

of specific target site variants (FRT, lox), have increased the

efficiency and scope of these tools (Turan et al., 2011). In addi-

tion to reporter lines where a single marker is activated on

recombination, switchable lines inwhich recombination removes

or inverses a first fluorescent reporter cassette, so that a second

cassette is expressed, permit marking of cell types before and

after recombination (Muzumdar et al., 2007 in mouse; Boniface

et al., 2009 in fish). In order to follow all cell derivatives a ubiqui-

tously expressed regulatory sequence controlling the conditional

reporter is required. In mouse, targeting to the Rosa26 locus is

frequently used, with an additional CAG promoter sequence to

398 Developmental Cell 21, September 13, 2011 ª2011 Elsevier Inc.

provide stronger and more ubiquitous

expression (Zong et al., 2005). In zebra-

fish, the ubiquitin promoter, chosen by

analogy with Drosophila, looks promising

(Mosimann et al., 2011).

Genetic manipulations that underlie

permanent cell labeling are spatially

controlled by the use of tissue-specific

promoters, to target a chosen progenitor

cell population. Mouse Cre lines are extensively used to follow

the descendants of cells that had expressed the Cre recombi-

nase. A repertoire ofCre lines,which include flexible locus target-

ing, continues to be developed by consortia such as EUCOMM.

This approach, described as ‘‘genetic inducible fate mapping,’’

was first employed in experiments where an Engrailed-Cre line

wascrossedwith ab-actin-loxSTOPlox-lacZ line to fatemapcells

originating at the mouse midbrain-hindbrain constriction (Zinyk

et al., 1998). In the fly model, the G-TRACE procedure (Evans

et al., 2009) is based on theGAL4-UAS binary expression system

(Brand and Perrimon, 1993), in which a sequence, encoding the

Flp recombinase, is under the control of UAS regulatory elements

that are targeted by the transcriptional activator GAL4, produced

from another transgene with tissue-specific regulatory elements

(Figures 2A and 2B). A strength of the Drosophila community

has been the large collection of UAS/Gal4 lines and further

resources, which integrate recombinase technology, is now

becoming available. The GAL4-UAS system is now also being

optimized for use in zebrafish, where lines are beginning to be

established. As an alternative to the use of a recombinase, the

Kaloop approach (Figure 2C) provides permanent cell labeling

by autoinduction of Gal4 under the control of UAS in the reporter

cassette independently of the tissue-specific promoter (Distel

et al., 2009).

In all these approaches, genetic tracing of progenitors labels

all the cells that had expressed the tissue-specific promoter

drivingCre, precluding any distinction between different progen-

itors, and should be interpreted as the identification of structures

that arise from a gene expression domain. Such genetic fate

mapping in mouse, frequently confused with lineage analysis,

Page 6: Developmental Cell Perspective - COnnecting REpositories · Developmental Cell Perspective Tracing Cells for Tracking Cell Lineage and Clonal Behavior Margaret E. Buckingham1, *and

Developmental Cell

Perspective

has given rise to controversial results and needs careful control;

transient gene expression in another progenitor cell population

or unexpected later expression in cells of the tissue under study

can confuse the analysis. This is exemplified by controversy over

an experiment on genetic tracing of Tbx18-positive epicardial

cells covering the mouse heart. In Tbx18-Cre;R26R embryos,

myocardial cells expressing the lacZ reporter were observed,

leading to the conclusion that the epicardium can give rise to

myocardium. This was challenged by another group that had

observed Tbx18 expression in some myocardial cells, which

would trigger transcription of the reporter independently of

expression in the epicardium (Christoffels et al., 2009). Transient

temporal activation of Cre/Flp recombinases reduces the

problem posed by misleading domains of expression.

Temporal regulation of marker gene expression is now an

important facet of genetic tracing. In Drosophila, this can be

achieved by activation, due to a change in temperature, of a

heat-shock promoter (hsp) regulating Flp (Harrison and Perri-

mon, 1993), or by the use of temperature-sensitive versions of

the GAL80 repressor (McGuire et al., 2003). In zebrafish, local-

ized, or indeed cell-specific, activation of a heat-shock promoter

has been successfully achieved by a laser-pointer-driven micro-

heater (Placinta et al., 2009). The MAZe system in zebrafish

(Figure 2D) depends on a self-excising hsp-Cre cassette that

then brings a GAL4-VP16 cassette under the control of a ubiqui-

tous promoter and leads to the activation of a UAS-driven fluo-

rescent reporter in the same transgene (Collins et al., 2010),

with resultant cell labeling. In mouse, regulation by temperature

changes is not possible, and inducible systems involving antibi-

otics, such as the Tet system (Gossen and Bujard, 1992), or

hormones, have been developed. The estrogen receptor (ER),

with tamoxifen as a ligand, is widely used, mainly in the Cre-

ERT2 version (Feil et al., 1997 in mouse; Mosimann et al., 2011

in fish). Tamoxifen administration by injection into the mother

usually results in recombination within 6–24 hr in the mouse

embryo (Hayashi and McMahon, 2002). In the case of the zebra-

fish embryo, immersion in a tamoxifen-containing medium,

which can be washed out, permits rapid removal of the ligand.

Imprecision in the timing of tamoxifen activation is a general

problem; an ingenious method has been described recently for

photoactivation of a caged form of tamoxifen in zebrafish, with

the added advantage that a single cell can be targeted, with

potential applications for clonal analysis (Sinha et al., 2010). As

discussed later, dose control to give a low frequency of random

recombination can be used to achieve clonal levels of cell

labeling, permitting lineage analysis.

Genetic tracing reflects the activity of a promoter. However,

mosaics, which are also based on genetic tools, provide wider

possibilities for bona fide cell labeling.

Mosaics: Simultaneous Labeling of Several ProgenitorsSimultaneous labeling of several progenitor cells may be useful

to assess the variability of cell fate potential or to recognize

differing cell fate choices after asymmetric division, or to monitor

cell dynamics. Mosaics correspond to the association of genet-

ically distinct cells, which reflect disparate, nonclonal labeling

of progenitor cells. Mosaic embryos have provided important

information about the origin of different cell types. Classic exper-

iments using allophenic (chimaeric) mice, derived from morulae

De

aggregation from different mouse lines, provided new insight

into the polyclonal origin of a tissue or structure from a small

number of founder cells, as exemplified by the melanocyte

lineage that determines the ordered patterning of stripes of

coat color (Mintz, 1965). Pioneering work based on the injection

of ES cells into the blastocyst to create chimaeras (Gardner and

Rossant, 1979) underlies more recent experiments. Thus, a

mixture of ES cells, each with the Rosa26 locus targeted to

express a distinct fluorescent protein, was introduced into wild-

type blastocysts. The resultant mosaic embryos were analyzed

to determine whether hematopoietic and endothelial cells in the

yolk sac blood islands arise from a common hemangioblast

progenitor. The authors concluded that each island had multiple

progenitors and that the contribution of a single hemangioblast to

both endothelial and hematopoietic lineages was a rare event

(Ueno and Weissman, 2006). Creation of mosaics followed

by time-lapse imaging has permitted cell tracking and fate

mapping, as illustrated by limited electroporation of plasmids

encoding H2B-EGFP and cytoplasmic DsRed into some cells of

the chick epiblast. Multiphoton time-lapse microscopy showed

the mechanism of the ‘‘polonaise’’ movements of cells that

precede gastrulation; in this case the mosaic of positive and

negative cells facilitated cell tracking (Voiculescu et al., 2007).

Genetic manipulations to produce mosaics have been classi-

cally used in Drosophila for clonal analysis. From pioneering

work by Sturtevant (1929) on gynandromorphs resulting from

spontaneous X-inactivation to current techniques for spatially

and temporally controlled activation of cell markers, the co-

herent growth of cells in the Drosophila embryo has facilitated

clonal analysis. Isolated clusters of labeled cells are generally

assumed to be clonal.

More sophisticated reporters to mosaic labeling continue to

be developed. For example, in mosaic analysis with double

markers (MADM) (Zong et al., 2005), Cre-mediated recombina-

tion between homologous mouse chromosomes results in the

generation of a complete coding sequence for GFP or RFP

from chimaeric sequences containing partial sequences of

the fluorescent proteins (Figures 3A and 3B). The use of split

sequences also underlies the twin-spot system in Drosophila

(Griffin et al., 2009). After mitosis, recombined chromatids may

segregate to mark daughter cells differently, such that cells

expressing either reporter come from a common progenitor.

With the development of spectral microscopy and of fluorescent

proteins with a wide range of emission spectra, the number of

markers that can be imaged simultaneously is increasing

(Nowotschin et al., 2009). The Brainbow system, developed

for mouse, depends on a stochastic choice between distinct

recombinase target sites flanking a range of fluorescent markers

in a transgene integrated in multiple copies. This leads to the

generation of a spectacular mosaic of differently colored cells

(Livet et al., 2007). With fluorescent proteins targeted to subcel-

lular compartments, as well as with recombinase-mediated

inversion of reporter sequences (Brainbow 2), the possible

combinations that can distinguish cells are huge (%90). Although

in the original paper the expression of the construct was

restricted to neuronal cell types, a universal rainbow line,

R26R-confetti (Snippert et al., 2010), coupled to existing specific

Cre lines, increases the range of applications. This was usedwith

a Cre-ERT2 under the control of Lgr5, which is expressed in stem

velopmental Cell 21, September 13, 2011 ª2011 Elsevier Inc. 399

Page 7: Developmental Cell Perspective - COnnecting REpositories · Developmental Cell Perspective Tracing Cells for Tracking Cell Lineage and Clonal Behavior Margaret E. Buckingham1, *and

C

mFRT71

EGFP mcherrymcitrine cerulean

mFRT71 mFRT71

1 - inversion 2 - inversion

3 - excision

UAS

D

E

m

l

heat shockhsp-mFlp5 1 2 3

Elav-Gal4155

N-GFP

GFP-CN-RFP

RFP-C

2- mitotic recombination1- nonmitotic recombination

A

1 2

N-GFP

GFP-CN-RFP

RFP-C

N-GFP RFP-C

GFP-CN-RFP

B

MADM or twin-spot

Flybow

hsp-Flp or tissue-Creor Cre-ER

Figure 3. Examples of Reporter Constructs for Mosaic Analyses(A) In the MADM system in mouse or in the twin-spot system in Drosophila, partial sequences for fluorescent proteins are separated by recombination sites (<),which, when exposed to recombinase, reconstitute a fluorescent reporter. If mitotic recombination takes place, green and red fluorescent reporters markseparate sister cells as a result of allelic segregation. After nonmitotic recombination, cells become yellow (Zong et al., 2005; Griffin et al., 2009).(B) Schematic illustration of cell labeling in the lineage.(C) In Drosophila, the Flybow construct (version FB1.1) of membrane tagged fluorescent reporters can be rearranged by the action of Flp on FRT sites.(D) Generation of mosaics by heat-shock induction of the Flp recombinase, which induces stochastic labeling of cells. Elav-Gal4155 drives expression of the UASconstruct in neurons. Prior to expression the potential fluorescent marker is indicated as an outline; full colored circles indicate expressing cells.(E) An example of a mosaic in an L3 larval optic lobe, showing labeled lineages of medulla (m) and lamina (l) neurons, in blue, red, or yellow, after transient heatshock at early larval stages. EGFP (green) is the default fluorescent protein, when Flp is not active. The merged image and the red channel (detecting red andyellow cells) are shown on the left and right, respectively (Hadjieconomou et al., 2011, adapted with permission).

Developmental Cell

Perspective

cells of the crypt of the mouse intestine. Tamoxifen induction of

Cre at different time points, followed bymathematical analysis of

cell patterns marked with the four randomly generated reporters,

led to conclusions about stem cell turnover without asymmetric

cell divisions. Stochastic adoption of stem or transit amplifying

cell fates depends on neutral competition between cells. Such

sophisticated mathematical analysis of clone distributions has

also been applied to other tissues, and a general theoretical

framework, which discriminates between patterns of long-term

clonal evolution for distinguishing three classes of stem cell

behavior, has been proposed (Klein and Simons, 2011). Adapta-

tions of this system have now been described for Drosophila—

d-Brainbow (Hampel et al., 2011) or Flybow (Hadjieconomou

et al., 2011)—in which cell labeling as a result of different

stochastic recombination events is linked to the UAS/GAL4

system to drive expression of the transgene reporter construct.

In the d-Brainbow application, epitope tagged, aswell as fluores-

cent proteins, were used, thus permitting both imaging and histo-

logical examination of fixed tissue. The Flybowsystemavoids the

problem of Cre toxicity by employing Flp-mediated inversion of

reporter sequences and has been used to address questions

about the formation of neural network architecture (Figures 3C–

3E). As in all mosaic analyses, the challenge is to sort out cells

400 Developmental Cell 21, September 13, 2011 ª2011 Elsevier Inc.

following the color code, which can be limited by the spectral

separation of different combinations of fluorescent proteins and

by the light microscopy resolution of subcellular localization.

Mosaics have opened the way to spectacular multicolor

labeling of cells and have given insight into the polyclonal origin

of tissues, their architecture, and the cell dynamics underlying

tissue growth. The approaches discussed above, which inte-

grate tissue-specific or temporal control, can also potentially

be extended to introduce recombinases under cell-cycle control.

Since they are generated genetically, an important application of

mosaic approaches is that they can be combined with functional

analyses, based on the use of mutated alleles.

Clonal Analysis and Gene FunctionIn order to relate clonal growth to gene function, mutant clones

can be generated in a wild-type background or clonal analysis

can be performed in a mutant background. Following the work

of C. Stern (1936) onmitotic recombination, Flp-dependent inter-

chromosomal recombination had been used in Drosophila to

generate mutant clones that no longer express a marker (e.g.,

Xu and Rubin, 1993). An adaptation to produce positively

marked clones, mosaic analysis with repressible cell marker

(MARCM) (Figure 4A), leads to segregation of the mutant allele

Page 8: Developmental Cell Perspective - COnnecting REpositories · Developmental Cell Perspective Tracing Cells for Tracking Cell Lineage and Clonal Behavior Margaret E. Buckingham1, *and

after mitotic recombination

A1 A2MARCM

Gal80

tub

XX

Gal80

after mitotic recombination

B1 B2Twin-spot MARCM

RFP-miR

GFP-miR

GFP-miR

RFP-miR

X

X

C1 C2

12

lacZ

lacZ

[UAS-GFP] [tub-Gal4]

XX

[UAS-GFP] [tub-Gal4] hsp-Flp

X

X

X

X

hsp-Flp

X

X

RFP-miR

RFP-miR

X

X

[GFP] [RFP]

GFP-miR

GFP-miR

[GFP] [RFP]

[GFP] [RFP]

Gal80

tub

XX

Gal80

[UAS-GFP] [tub-Gal4]

X

hsp-Flp

X

hsp-Flp

1

2 X

X

X

X

X X

ubiquitous

Figure 4. Schematic Representations of Different Approaches to Tracing Cells in Mutant Clones in Drosophila(A1) The MARCM system depends on the elimination, by mitotic recombination, of the Gal80 transgene, which lies on the same chromosome arm as the mutantallele (X). This results in their segregation and thus the activation of the GFP reporter in the homozygote mutant cells (5).(A2) Transient heat-shock activation of the Flp recombinase results in reporter expression in mutant cells (Lee and Luo, 1999).(B1 and B2) In twin-spotMARCM, the two systems are combined (see also Figure 3A), in this case usingmicroRNAs (miR) as repressors ofGFP orRFP expression(Yu et al., 2009).(C1 and C2) To trace lineage progression in mutant clones, the MARCM system of GFP activation is combined with the system of heritable expression of lacZ(Harrison and Perrimon, 1993). A second reporter (lacZ) is activated after a second heat-shock-induced recombination that brings it under the control ofa ubiquitous promoter (Perdigoto et al., 2011). <: FRT sites for Flp recombination.

Developmental Cell

Perspective

from the repressor GAL80, so that a UAS-driven fluorescent

reporter is now activated in a mutated daughter cell and its

descendants (Lee and Luo, 1999). Twin-spot MARCM combines

the two approaches to follow sister cells (Yu et al., 2009). In this

case, repressors are microRNAs that target UAS-dependent

markers and are lost after mitotic recombination (Figure 4B).

This reduces the delay of MARCM derepression, which other-

De

wise depends on GAL80 decay. A potential difficulty is to deter-

mine when the mutation becomes effective, depending on the

perdurance of the endogenous protein. Ideally, one would like

to study lineage progression within clones of mutant cells. This

can be achieved inDrosophila by combining theMARCMsystem

with an independent Flp-induced recombination event that acti-

vates expression of a lacZ reporter (Figure 4C), so that two

velopmental Cell 21, September 13, 2011 ª2011 Elsevier Inc. 401

Page 9: Developmental Cell Perspective - COnnecting REpositories · Developmental Cell Perspective Tracing Cells for Tracking Cell Lineage and Clonal Behavior Margaret E. Buckingham1, *and

Developmental Cell

Perspective

sequential recombination events can generate clones of

b-galactosidase-positive cells within GFP marked clones of

mutant cells (Perdigoto et al., 2011). In this way, Notch signaling

in the lineage progression of intestinal stem cells has been

shown to restrict their self-renewal as well as affecting the later

stage of terminal differentiation.

Mosaics of cells with distinct genotypes also provide insight

into cell-cell interactions and the importance of cell competition

in selecting the progenitor of tissues and organs (Morata and

Ripoll, 1975). For tracing the descendants of mutant cells in

heterozygote mice, the MADM system can be used with distinct

fluorescent reporters for wild-type and mutant alleles. For this

purpose, a new reporter line, MADM-11, has been developed

to generate clones mutant for a gene located on chromosome

11. With a Cre recombinase under the control of Emx1, which

is expressed in cortical progenitors of the forebrain, it was shown

that components of the Lis1/Ndel1/14.3.3ε complex, which is

defective in lissencephaly syndromes, have distinct cell-autono-

mous functions during different stages of neuronal migration

(Hippenmeyer et al., 2010). The approach in which differently

marked ES cells are introduced into amouse blastocyst to create

mosaics can be combined with functional studies on mutant

ES cells to investigate the cell-autonomous roles of a gene. Alter-

natively, with a specific Cre, genetic tracing of clones can be

achieved. Using a Flk1-Cre integrated into the genome of the

reporter ES cells it was shown that all the endothelial cell

derivatives in the blood islands were derived from progenitors

expressing robust levels of Flk1, whereas most hematopoietic

cells were not (Ueno and Weissman, 2006). Mosaics are also

employed to perform clonal analysis on a mouse mutant back-

ground. Experiments in which insertion of a GFP reporter into

the X chromosome resulted in random X-inactivation made it

possible to follow cell dynamics in the developing limb bud by

live imaging in a Wnt5a mutant embryo (Gros et al., 2010).

Classic single-cell microinjection into the mouse epiblast has

also been carried out with mutant embryos, for example to

show that Otx2 is not required for proliferation of the visceral

endoderm lineage but is essential for anteriorly directed cell

movement (Perea-Gomez et al., 2001).

In addition to examining the effects of mutations on cell fate

choice and associated cell behavior, a related challenge is to

integrate the cellular data with dynamic gene expression, to

understand how genes are expressed or repressed during

progression along a lineage tree. The use of reporters of gene

expression with limited stability, such as the fusion protein

H2B-GFP (Plusa et al., 2008) or destabilized GFP (Harper et al.,

2010), permits reliable imaging of gene expression dynamics.

Integrating quantitative data on transcription factor kinetics

with subsequent lineage patterning is now realizable. Thus

monitoring the nucleocytoplasmic movement of Oct4 fused to

a photoactivatable GFP has demonstrated that differences in

Oct4 kinetics predict the future identity of mouse blastocyst

lineages (Plachta et al., 2011).

Modeling the emergence of different cell types in a lineage is

an emerging theme, as, for example, in C. elegans, for which

a complete lineage tree has been reconstructed and regulatory

genes have been identified. Such a predictive model gives an

indication of the number of regulatory factors required for reca-

pitulating the lineage, the synergistic variation of factors, and

402 Developmental Cell 21, September 13, 2011 ª2011 Elsevier Inc.

where, in the cell lineage tree, asymmetry might be controlled

by external influences (Larsson et al., 2011). The superposition

of experimentally determined gene regulatory networks on cell

lineage is beautifully illustrated by pioneering work on the sea

urchin embryo, where lineage is mainly invariant and early

lineage segregation has been examined on a cell-by-cell basis

in terms of transcriptional regulation and cell signaling. In this

way, for example, the endoderm gene regulatory network has

been defined up to the midblastula stage, giving new insight

also into the progressive segregation of endodermal from

mesodermal lineages (Peter and Davidson, 2010).

In most of the mosaic analyses previously discussed, the

lineage is preidentified by the use of tissue-specific regulatory

sequences and in this respect is therefore prospective. The inter-

pretation of mosaics in terms of clones is limited to local events

when growth is coherent and clusters of cells can be considered

as clonal units. In retrospective clonal analysis, it is possible to

lower the frequency of cell labeling to reach clonal conclusions,

even when growth is dispersive and to do this on the scale of the

whole organism.

Retrospective Clonal Analysis: Systematic Analysisof All Progenitors of a StructureProspective lineage analysis depends on a preconceived idea

about the progenitor cell population. Preidentification of the

potential stage and location of the progenitors to label is

required. However, this is not always known and potentially

restricts conclusions on lineage by not considering other coun-

terintuitive options. In contrast, retrospective approaches to

cell lineage depend on analyzing labeled cells at the end point

of the experiment and deducing their interrelationships and

previous history. Retrospective clonal analysis based on the

random genetic labeling of progenitors at a low frequency

constitutes a progenitor screen and permits the systematic anal-

ysis of the potential of any progenitor to colonize a particular

structure. We distinguish two kinds of retrospective clonal

analyses, depending on whether labeling is random in space,

but with temporal control, or random in both space and time.

Spatially Random Labeling

Clones that are induced at a particular time but result from

random spatial labeling have been produced in Drosophila by

X-irradiation induced recombination, pioneered by H.J. Becker

(1957), and subsequently employed to generate, in a heterozy-

gous Minute mutant background, Minute-positive cells with

a cuticular marker, which tend to outgrow their mutant neighbors

(Garcia-Bellido et al., 1973). The distribution of clusters of such

cells demonstrated the existence of internal demarcation lines

in the wing disc, which the clones did not cross. This classic

work led to important concepts and definitions, including

those of clonal compartments and clonal boundaries (Garcia-

Bellido, 1985). In the plant kingdom, elegant experiments

on genetic variegation have manipulated transposon-mediated

gene silencing. For example, a change in temperature led to

low-frequency mobility of a transposon in the promoter region

of the Pal gene, which encodes a red pigment, resulting in

restoration of gene function to give red sectors (clones) on the

ivory background of an Antirrhinum petal. In such experiments,

the sequence of lineage restrictions in the developing floral

meristem has been revealed (Vincent et al., 1995). Sophisticated

Page 10: Developmental Cell Perspective - COnnecting REpositories · Developmental Cell Perspective Tracing Cells for Tracking Cell Lineage and Clonal Behavior Margaret E. Buckingham1, *and

Developmental Cell

Perspective

quantitative analysis of sector parameters coupled to computer

modeling led to conclusions on the growth parameters that

are essential to give shape to the flower petal (Rolland-Lagan

et al., 2005).

In amniotes, infection with replication-defective retroviruses

that integrate into the genome provides a spatially random

labeling of progenitors at a defined time. This approach, first

described for themouse (Saneset al., 1986),was refined for clonal

analysis by the development of libraries of individually marked

retroviruses, where each member encodes a reporter and has

a DNA tag (Golden et al., 1995). The complexity of the library

permits evaluation of clonality between labeled cells, based on

the presence of the same tag, identified by PCR. This approach

has been extensively used in birds and mammals, especially for

characterizing lineage segregation in the central nervous system,

as in the early demonstration that clonal derivatives contribute to

more than onemajor subdivision of the telencephalon (Walsh and

Cepko, 1992). In a recent adaptation, used for clonal analysis of

blood cell types, infection with a barcoded retroviral library

carrying a fluorescent marker was followed by separation of indi-

vidual circulating cells by flow cytometry and sensitive sequence-

based characterization of clonally related cells (Gerrits et al.,

2010). Potential problems for random retroviral labeling are inte-

gration site effects and variable infectivity, illustrated by murine

retroviruses that only infect proliferative cells, a limitation partially

overcome by lentiviruses based vectors. As an alternative to

libraries, low-level infection of GFP encoding retro- and lentivi-

ruses has been used to mark single cells—for example, recently

in the zebrafish brain, to show that whereas neuroblasts undergo

a limited amplification, single radial glial cells self-renew and

generatedifferent cell types, thusbehavingasbonafidestemcells

in vivo (Rothenaigner et al., 2011).

Inducible recombinase systems based on the use of a ubiqui-

tous reporter also provide random spatial labeling of progeni-

tors. Adjusting the duration and temperature difference of the

heat shock or the dose of tamoxifen can result in control of

the frequency of labeling, to permit clonal conclusions. This

approach (Figures 5A–5D), in which low doses of tamoxifen

were administered to a CMV-CreERT2;R26R mouse line, was

instrumental in showing the organization of stem cells in the

matrix of the hair follicle and the mode of growth of their deriva-

tives (Legue and Nicolas, 2005). In this example, statistical anal-

ysis was necessary to assess the probability of independent

labeling events and to conclude on the clonal relationship

between two labeled groups of cells. In a refinement of this

method, two inducible reporters, R26R (lacZ) and R26R-EYFP,

were used together to help to distinguish clonal events (Arques

et al., 2007). Sophisticated quantitative analysis of such clonal

patterns, coupled to computer modeling, has shed new light

on the mechanism of limb bud growth (Marcon et al., 2011).

Retrospective examination of lineage in this way, by activation

of reporter expression at different time points, extends the

potential for precise temporal reconstruction of lineage trees.

Spatially and Temporally Random Labeling

Other approaches to retrospective clonal analysis are random in

both space and time and therefore encompass the complete

history of a lineage. The accumulation of random somatic cell

mutations during normal development provides an endogenous

marker of cell lineage. Thus, analysis of mutations in microsatel-

De

lite DNAs, at the single-cell level, using the new sequencing

technologies, has led to the construction of mammalian lineage

trees for a number of tissue types with easily isolated cells, such

as the blood (Wasserstrom et al., 2008). This method is labor

intensive and requires sophisticated computational analyses

but is noninvasive, with the advantage that it is also applicable

to human material.

Another approach depends on the introduction, as a transgene

or targeted to an endogenous locus, of a nlaacZ reporter

sequence, rendered nonfunctional by a duplication that intro-

duces a STOP codon into the b-galactosidase coding sequence.

A rare, random event of intragenic recombination will generate

a functional nlacZ reporter, which is then transmitted genetically

to the descendants of the cell. This results in clonally related

labeled cells, which are detectable when the recombined nlacZ

lineage tracer is expressed (Figures 5E and 5F). The choice of

regulatory sequences controlling reporter expression deter-

mines the tissue analyzed at the end point but does not condition

the genetic labeling of the progenitor cells that give rise to it. The

rarity of the event makes clonal analysis possible (Bonnerot and

Nicolas, 1993). Collections of embryos are generated, in which

the frequency of labeling, in the structure under consideration,

is determined. To establish clonality, statistical analyses are

required, based on the frequency of observations, as, for

example, the fluctuation test of Luria and Delbruck (1943), which

estimates the probability of one or more than one recombination

events. The different types of clones that result from random

labeling can be divided into groups based on their characteris-

tics such as size, spatial distribution, and cell type. When similar

clones are observed it can be assumed that the library of clones

has reached saturation. From the collection of clones, derived

from progenitors that have undergone recombination at different

stages, the temporal history can be reconstructed, based on the

premise that subclones have more restricted cell fate potential

than parental clones. In addition to reconstruction of the lineage,

its number of founder cells, and diversification into sublineages,

important aspects of cell behavior such as the formation of clonal

boundaries, or asymmetric stem cell versus symmetric prolifera-

tive modes of cell division, can also be deduced from the prop-

erties of clones within the library (Nicolas et al., 1996; Petit et al.,

2005). An example is provided by analysis of clones in embryos

of an a-cardiac actinnlaacZ/+ mouse line, which led to the demon-

stration of two myocardial cell lineages, which segregate early,

with distinct and overlapping contributions to different parts of

the heart (Meilhac et al., 2004). Subsequent analysis, using the

same mouse line, established sublineages, within the second

myocardial lineage, that contribute to different parts of the arte-

rial pole of the heart and also to different skeletal muscle groups

in the head (Lescroart et al., 2010) (Figures 5G–5L). The nlaacZ

approach has resulted in new lineage insights for many tissues

in the mouse, including segregation of the germ layers during

gastrulation (Tzouanacou et al., 2009), and should be applicable

to other species. A similar reporter, based on duplication in the

b-glucuronidase gene, has been used for clonal analysis in plants

(Swoboda et al., 1994).

The strength of random retrospective clonal analyses is to

reconstruct lineage trees over an extended time scale and to

understand the mode of regionalization of a structure, by target-

ing systematically all progenitors. However, the inference of the

velopmental Cell 21, September 13, 2011 ª2011 Elsevier Inc. 403

Page 11: Developmental Cell Perspective - COnnecting REpositories · Developmental Cell Perspective Tracing Cells for Tracking Cell Lineage and Clonal Behavior Margaret E. Buckingham1, *and

A

lacZSTOP

ROSA26

low dose of tamoxifen,Cre-ERT2

B

C

E14.5-2084

OFT

E14.5-2901RV

J

1BA2BA

1

3

67

24

5

OFT

RV

1BA

heart head

nlacZ

β-gal

nlaacZ

2

3

56

4

E8.5-156

1

25

6

E8.5-204

first lineage

second lineage

K L

EG H I

F

α-cardiac actin

STOP

α-cardiac actin

spontaneous recombination

nlacZ

nlaacZ

D

loxP

medulla clonecuticle cloneIRS clone

2BA

Figure 5. Retrospective Clonal Analysis in Mouse(A–D) Inducible clonal analysis based on recombination of a conditional ROSA26 reporter (A) activated by low doses of tamoxifen (B). Clones in different layers(color coded; IRS, inner root sheath) of the hair follicle originate from different domains (red arrowheads) of the matrix which is a source of stem cells (C and D).(Legue and Nicolas, 2005, adapted with permission).(E–L) Random retrospective clonal analysis by the laacZ approach (E) The laacZ reporter is rendered nonfunctional by an internal duplication, which canspontaneously recombine into a functional lacZ gene, at a low frequency.(F) Random generation of an lacZ-positive clone (cells outlined in blue), which is detectable in the expression domain of the ac-actin promoter, i.e., in cardiac andskeletal muscles (full blue circles indicate b-galactosidase [b-gal]-positive cells).(G and H) Examples of b-gal-positive clones with an exclusive contribution to region 1 (outflow tract) or 3 (left ventricle) of the myocardium, indicative of lineagesegregation. E8.5, embryonic day 8.5 (followed by clone number).(I) The contributions of the first and secondmyocardial lineages, based on the analysis of 3,629 embryonic heart tubes, are summarized in red and green (Meilhacet al., 2004).(J and K) Examples of b-gal-positive clones of the second myocardial lineage colonizing both skeletal muscles of the head and myocardium of the heart, takenfrom a collection of 2,223 fetuses.(L) The lineage contributing to headmuscles derived from the first branchial arch (1BA), which also contributes to the right ventricle (RV), is shown in blue, while thelineage that contributes to second branchial arch-derived head muscles (2BA) and to the outflow tract (OFT) is represented in pink (Lescroart et al., 2010).

Developmental Cell

Perspective

events that precede the observations can be controversial. To

gain direct access to the dynamics of lineage progression, live

analyses are required, associated with successive observations

at shorter time intervals.

404 Developmental Cell 21, September 13, 2011 ª2011 Elsevier Inc.

Four-Dimensional Imaging of Lineage ProgressionA grail of lineage analysis is the complete four-dimensional (4D)

imaging of cells in vivo. This was pioneered in the chick, in which

time-lapse imaging was first attempted over 80 years ago

Page 12: Developmental Cell Perspective - COnnecting REpositories · Developmental Cell Perspective Tracing Cells for Tracking Cell Lineage and Clonal Behavior Margaret E. Buckingham1, *and

Figure 6. Lineage Reconstruction for the Early Zebrafish EmbryoBased on 4D Imaging(A–C) Images of mitotic spindles by second harmonic generation (SHG) signal(A), of chromosomes marked by H2B-mcherry (B) and membranes by thirdharmonic generation (THG) signal (C).(D1 and D2) Digital reconstruction of the embryos with color-coded cell line-ages at the 8-cell (D1) and 512-cell (D2) stages.(E) An example of information on cell behavior resulting from this approachshows the orientations of cell divisions, where each color corresponds tosuccessive cell cycles (Olivier et al., 2010, adapted with permission).

Developmental Cell

Perspective

(Wetzel, 1929). A complete lineage tree has been accomplished

for C. elegans using Nomarski optics and direct observation to

track cells (Sulston et al., 1983). This remarkable achievement

was rendered possible by the transparency and small size (fewer

than 1,000 cells) of the nematode. Furthermore, the fact that the

lineage was found to be invariant facilitated the analysis. Inter-

estingly, in spite of the fixed relationship between cell ancestry

and cell fate, the correlation between them lacks obvious

pattern; for example, neurons do not all derive from ectoderm

and only intestinal and germ cells are of monoclonal origin.

With fluorescent markers facilitating in vivo imaging, as dis-

cussed in previous sections, tracking labeled cells has led to

information on cell fate and associated behavior in many organ-

isms. In classic experiments in zebrafish, partial lineages were

reconstructed by video recording, leading, for example, to the

demonstration that specific cell behavior is coupled to particular

cell cycles and appears to account for clonal restriction in neural

De

cell fate (Kimmel et al., 1994). Time-lapse imaging of cortical sli-

ces in the mouse embryo, after in utero infection with GFP-ex-

pressing retroviruses, showed that radial glia generate neurons

by asymmetric cell division (Noctor et al., 2001). However, with

the exception of experiments on invertebrates, only fragmentary

information on specific lineages has been obtained, mainly

limited by the cell-labeling procedure.

For more systematic lineage reconstructions, pioneering

experiments in plants, where the absence of cell migration and

also of apoptosis facilitate cell tracking, have used fluorescent

markers expressed in all cells, with a subcellular resolution,

targeting the membrane or chromatin or revealing cell-cycle

stages. In this case, confocal microscopy with long-term

(12 days), as well as short-term, imaging was employed to

analyze the development of the flower primordium from the

meristem in Arabidopsis. Image registration algorithms were

developed to assist lineage reconstruction (Reddy et al., 2004).

In the plant field, the challenge of quantitative analysis of growth

parameters over time is being met by new tools for image pro-

cessing and reconstruction to track cell lineages (Fernandez

et al., 2010). During animal development, confocal and multi-

photon microscopy are currently extensively used for imaging

cells. However, problems of light scattering and resolution are

particularly critical for cell tracking. With the current interest in

stem cells in the adult, as well as during development, accessing

cells that are located deep within an organism can be a major

problem. Advances in light sheet microscopy (e.g., SPIM,

DSLM), successfully used on zebrafish embryos at early (Keller

et al., 2008) or late (Swoger et al., 2011) developmental stages,

hold out new promise for minimally invasive, high-resolution

images with good penetration depth and fast acquisition

(Huisken and Stainier, 2009). By following fluorescently labeled

nuclei, with an automated image segmentation procedure, Keller

et al. (2008) provide a resource of ‘‘digital embryos,’’ for lineage

analysis over 24 hr, from early cleavage stages until the onset of

organogenesis. Another new development, which is based on

label-free multiphoton technology with spiral scanning to opti-

mize resolution, penetration, and photoperturbation, has led to

complete lineage reconstruction of zebrafish early development,

up until the 1,000-cell (blastula) stage (Olivier et al., 2010)

(Figure 6). This remarkable technical feat exploited the intrinsic

optical nonlinear properties of the sample (harmonic genera-

tion)—namely, the oriented microtubules of the spindle and

aqueous/lipidic surfaces such as membranes—together with

two-photon excitation of a fluorescent chromosomal marker en-

coded by a transgene. This analysis required sophisticated

processes of image acquisition, as well as algorithms for auto-

mated lineage reconstruction, which nevertheless still relied on

time-consuming visual verification. A challenge for these new

approaches is the requirement for higher-resolution multicolor

cell imaging and for optimization of algorithms for fast and auto-

mated image reconstruction in dense environments, so as to

allow unambiguous cell tracking. A practical problem for extend-

ing long-term imaging to later stages is the need to immobilize

the animal without impeding development. It is not clear how

far 4D analysis will progress to map cell behavior from the outset

of development, but it also provides the potential to extend

fragmentary imaging of later cell lineage choices into more

comprehensive documentary films of cell history.

velopmental Cell 21, September 13, 2011 ª2011 Elsevier Inc. 405

Page 13: Developmental Cell Perspective - COnnecting REpositories · Developmental Cell Perspective Tracing Cells for Tracking Cell Lineage and Clonal Behavior Margaret E. Buckingham1, *and

Developmental Cell

Perspective

In conclusion, the technological developments discussed here

have opened up new horizons. Genetic tools, which are

becoming available for an increasing number of organisms,

have solved the old problem of marker dilution at cell division

and have introduced sophisticated methods for spatio-temporal

targeting of labeling. Developments in the range of fluorescent

markers now permit direct and multicolor observations, which

can be orchestrated at will by genetic engineering. The classical

problem of clonality of the labeling is addressed either by

combining several markers or by lowering the frequency of

labeling. Advances in microscopy are crucial for recording clonal

data with increasing resolution in four dimensions. Sophisticated

computational methods are being developed to analyze the

large data sets generated by clonal analyses and to provide

in-depth understanding of the cellular mechanisms leading to

the observed clonal patterns. A challenge for the next decade

is to grasp the significance of changes in cell behavior followed

at a single-cell level and to integrate the cellular with the

molecular dimension to understand lineage choices and lineage

progression. In the future, in vivo imaging and genetic manip-

ulation of markers will be widely applicable to the diversity

of species already apprehended in Evo/Devo type studies, no

doubt leading to unexpected conceptual lineage developments

and revealing the cellular aspects of evolutionary ‘‘tinkering’’

(Jacob, 1977).

ACKNOWLEDGMENTS

We thank P. Herbomel, E. Hirsinger, J.-F. Nicolas, and F. Schweisguth forinsightful discussions and anonymous reviewers for comments. Work onlineage analysis in the Buckingham lab is supported by the Institut Pasteur,the CNRS (URA 2578) and by the EU through the CardioCell (FP7 - HEALTH-2007-2.4.2-5) project. S.M.M. is an INSERM research scientist.

REFERENCES

Arques, C.G., Doohan, R., Sharpe, J., and Torres, M. (2007). Cell tracingreveals a dorsoventral lineage restriction plane in the mouse limb bud mesen-chyme. Development 134, 3713–3722.

Baker, S.M., Buckheit, R.W., 3rd, and Falk, M.M. (2010). Green-to-redphotoconvertible fluorescent proteins: tracking cell and protein dynamics onstandard wide-field mercury arc-based microscopes. BMC Cell Biol. 11, 15.

Ba1akier, H., and Pedersen, R.A. (1982). Allocation of cells to inner cell massand trophectoderm lineages in preimplantation mouse embryos. Dev. Biol.90, 352–362.

Becker, H.J. (1957). Roentgen mosaic spots & defective mutations at theeye of Drosophila & the evolutive physiology of the eye. Z. Indukt. Abstamm.Vererbungsl. 88, 333–373.

Beddington, S.P. (1981). An autoradiographic analysis of the potency ofembryonic ectoderm in the 8th day postimplantation mouse embryo. J. Em-bryol. Exp. Morphol. 64, 87–104.

Ben-Yair, R., and Kalcheim, C. (2005). Lineage analysis of the avian dermo-myotome sheet reveals the existence of single cells with both dermal andmuscle progenitor fates. Development 132, 689–701.

Boniface, E.J., Lu, J., Victoroff, T., Zhu, M., and Chen, W. (2009). FlEx-basedtransgenic reporter lines for visualization of Cre and Flp activity in live zebra-fish. Genesis 47, 484–491.

Bonnerot, C., and Nicolas, J.F. (1993). Clonal analysis in the intact mouseembryo by intragenic homologous recombination. C. R. Acad. Sci. III 316,1207–1217.

406 Developmental Cell 21, September 13, 2011 ª2011 Elsevier Inc.

Brand, A.H., and Perrimon, N. (1993). Targeted gene expression as ameans ofaltering cell fates and generating dominant phenotypes. Development 118,401–415.

Caron, S.J., Prober, D., Choy, M., and Schier, A.F. (2008). In vivo birthdating byBAPTISM reveals that trigeminal sensory neuron diversity depends on earlyneurogenesis. Development 135, 3259–3269.

Christian, M., Cermak, T., Doyle, E.L., Schmidt, C., Zhang, F., Hummel, A.,Bogdanove, A.J., and Voytas, D.F. (2010). Targeting DNA double-strandbreaks with TAL effector nucleases. Genetics 186, 757–761.

Christoffels, V.M., Grieskamp, T., Norden, J., Mommersteeg, M.T., Rudat, C.,and Kispert, A. (2009). Tbx18 and the fate of epicardial progenitors. Nature458, E8–E9, discussion E9–E10.

Cockburn, K., and Rossant, J. (2010). Making the blastocyst: lessons from themouse. J. Clin. Invest. 120, 995–1003.

Collins, R.T., Linker, C., and Lewis, J. (2010). MAZe: a tool for mosaic analysisof gene function in zebrafish. Nat. Methods 7, 219–223.

Conklin, E.G. (1905). The organization and cell lineage of the ascidian egg. J.Acad. Nat. Sci. Phila. 13, 1–119.

Distel, M., Wullimann, M.F., and Koster, R.W. (2009). Optimized Gal4 geneticsfor permanent gene expression mapping in zebrafish. Proc. Natl. Acad. Sci.USA 106, 13365–13370.

Evans, C.J., Olson, J.M., Ngo, K.T., Kim, E., Lee, N.E., Kuoy, E., Patananan,A.N., Sitz, D., Tran, P., Do, M.T., et al. (2009). G-TRACE: rapid Gal4-basedcell lineage analysis in Drosophila. Nat. Methods 6, 603–605.

Feil, R., Wagner, J., Metzger, D., and Chambon, P. (1997). Regulation of Crerecombinase activity by mutated estrogen receptor ligand-binding domains.Biochem. Biophys. Res. Commun. 237, 752–757.

Fernandez, R., Das, P., Mirabet, V., Moscardi, E., Traas, J., Verdeil, J.L., Ma-landain, G., and Godin, C. (2010). Imaging plant growth in 4D: robust tissuereconstruction and lineaging at cell resolution. Nat. Methods 7, 547–553.

Galli, D., Domınguez, J.N., Zaffran, S., Munk, A., Brown, N.A., and Bucking-ham, M.E. (2008). Atrial myocardium derives from the posterior region of thesecond heart field, which acquires left-right identity as Pitx2c is expressed.Development 135, 1157–1167.

Galperin, C. (1998). From cell lineage to developmental genetics. Hist. Philos.Life Sci. 20, 301–344.

Garcia-Bellido, A. (1985). Cell lineages and genes. Philos. Trans. R. Soc. Lond.B Biol. Sci. 312, 101–128.

Garcia-Bellido, A., Ripoll, P., and Morata, G. (1973). Developmental compart-mentalisation of the wing disk of Drosophila. Nat. New Biol. 245, 251–253.

Gardner, R.L., and Rossant, J. (1979). Investigation of the fate of 4-5 day post-coitum mouse inner cell mass cells by blastocyst injection. J. Embryol. Exp.Morphol. 52, 141–152.

Gerrits, A., Dykstra, B., Kalmykowa, O.J., Klauke, K., Verovskaya, E., Broe-khuis, M.J., de Haan, G., and Bystrykh, L.V. (2010). Cellular barcoding toolfor clonal analysis in the hematopoietic system. Blood 115, 2610–2618.

Gline, S.E., Kuo, D.H., Stolfi, A., and Weisblat, D.A. (2009). High resolutioncell lineage tracing reveals developmental variability in leech. Dev. Dyn. 238,3139–3151.

Golden, J.A., Fields-Berry, S.C., and Cepko, C.L. (1995). Construction andcharacterization of a highly complex retroviral library for lineage analysis.Proc. Natl. Acad. Sci. USA 92, 5704–5708.

Gossen, M., and Bujard, H. (1992). Tight control of gene expression inmammalian cells by tetracycline-responsive promoters. Proc. Natl. Acad.Sci. USA 89, 5547–5551.

Griffin, R., Sustar, A., Bonvin, M., Binari, R., del Valle Rodriguez, A., Hohl, A.M.,Bateman, J.R., Villalta, C., Heffern, E., Grunwald, D., et al. (2009). The twin spotgenerator for differential Drosophila lineage analysis. Nat. Methods 6,600–602.

Gros, J., Hu, J.K., Vinegoni, C., Feruglio, P.F., Weissleder, R., and Tabin, C.J.(2010). WNT5A/JNK and FGF/MAPK pathways regulate the cellular eventsshaping the vertebrate limb bud. Curr. Biol. 20, 1993–2002.

Page 14: Developmental Cell Perspective - COnnecting REpositories · Developmental Cell Perspective Tracing Cells for Tracking Cell Lineage and Clonal Behavior Margaret E. Buckingham1, *and

Developmental Cell

Perspective

Hadjieconomou, D., Rotkopf, S., Alexandre, C., Bell, D.M., Dickson, B.J., andSalecker, I. (2011). Flybow: genetic multicolor cell labeling for neural circuitanalysis in Drosophila melanogaster. Nat. Methods 8, 260–266.

Hampel, S., Chung, P., McKellar, C.E., Hall, D., Looger, L.L., and Simpson,J.H. (2011). Drosophila Brainbow: a recombinase-based fluorescence labelingtechnique to subdivide neural expression patterns. Nat. Methods 8, 253–259.

Harper, C.V., Featherstone, K., Semprini, S., Friedrichsen, S., McNeilly, J.,Paszek, P., Spiller, D.G., McNeilly, A.S., Mullins, J.J., Davis, J.R., and White,M.R. (2010). Dynamic organisation of prolactin gene expression in livingpituitary tissue. J. Cell Sci. 123, 424–430.

Harrison, D.A., and Perrimon, N. (1993). Simple and efficient generation ofmarked clones in Drosophila. Curr. Biol. 3, 424–433.

Hatta, K., Tsujii, H., and Omura, T. (2006). Cell tracking using a photoconvert-ible fluorescent protein. Nat. Protoc. 1, 960–967.

Hayashi, S., and McMahon, A.P. (2002). Efficient recombination in diversetissues by a tamoxifen-inducible form of Cre: a tool for temporally regulatedgene activation/inactivation in the mouse. Dev. Biol. 244, 305–318.

Hippenmeyer, S., Youn, Y.H., Moon, H.M., Miyamichi, K., Zong, H., Wynshaw-Boris, A., and Luo, L. (2010). Genetic mosaic dissection of Lis1 and Ndel1 inneuronal migration. Neuron 68, 695–709.

Hirose, G., and Jacobson, M. (1979). Clonal organization of the central nervoussystem of the frog. I. Clones stemming from individual blastomeres of the16-cell and earlier stages. Dev. Biol. 71, 191–202.

Huisken, J., and Stainier, D.Y. (2009). Selective plane illumination microscopytechniques in developmental biology. Development 136, 1963–1975.

Jacob, F. (1977). Evolution and tinkering. Science 196, 1161–1166.

Keller, P.J., Schmidt, A.D., Wittbrodt, J., and Stelzer, E.H. (2008). Recon-struction of zebrafish early embryonic development by scanned light sheetmicroscopy. Science 322, 1065–1069.

Kimmel, C.B., and Law, R.D. (1985a). Cell lineage of zebrafish blastomeres.I. Cleavage pattern and cytoplasmic bridges between cells. Dev. Biol. 108,78–85.

Kimmel, C.B., and Law, R.D. (1985b). Cell lineage of zebrafish blastomeres. III.Clonal analyses of the blastula and gastrula stages. Dev. Biol. 108, 94–101.

Kimmel, C.B., and Warga, R.M. (1986). Tissue-specific cell lineages originatein the gastrula of the zebrafish. Science 231, 365–368.

Kimmel, C.B., Warga, R.M., and Kane, D.A. (1994). Cell cycles and clonalstrings during formation of the zebrafish central nervous system. Development120, 265–276.

Klein, A.M., and Simons, B.D. (2011). Universal patterns of stem cell fate incycling adult tissues. Development 138, 3103–3111.

Larsson, J.K., Wadstromer, N., Hermanson, O., Lendahl, U., and Forchheimer,R. (2011). Modelling cell lineage using a meta-Boolean tree model witha relation to gene regulatory networks. J. Theor. Biol. 268, 62–76.

Lawson, K.A., Meneses, J.J., and Pedersen, R.A. (1991). Clonal analysis ofepiblast fate during germ layer formation in the mouse embryo. Development113, 891–911.

Le Douarin, N., and Barq, G. (1969). Use of Japanese quail cells as ‘‘biologicalmarkers’’ in experimental embryology. C. R. Acad. Sci. Hebd. Seances Acad.Sci. D 269, 1543–1546.

Lee, T., and Luo, L. (1999). Mosaic analysis with a repressible cell marker forstudies of gene function in neuronal morphogenesis. Neuron 22, 451–461.

Legue, E., and Nicolas, J.F. (2005). Hair follicle renewal: organization ofstem cells in the matrix and the role of stereotyped lineages and behaviors.Development 132, 4143–4154.

Lescroart, F., Kelly, R.G., Le Garrec, J.F., Nicolas, J.F., Meilhac, S.M., andBuckingham, M. (2010). Clonal analysis reveals common lineage relationshipsbetween head muscles and second heart field derivatives in the mouseembryo. Development 137, 3269–3279.

De

Livet, J., Weissman, T.A., Kang, H., Draft, R.W., Lu, J., Bennis, R.A., Sanes,J.R., and Lichtman, J.W. (2007). Transgenic strategies for combinatorialexpression of fluorescent proteins in the nervous system. Nature 450, 56–62.

Luria, S.E., andDelbruck,M. (1943). Mutations of bacteria from virus sensitivityto virus resistance. Genetics 28, 491–511.

Marcon, L., Arques, C.G., Torres, M.S., and Sharpe, J. (2011). A computationalclonal analysis of the developing mouse limb bud. PLoS Comput. Biol. 7,e1001071.

McCauley, D.W., and Bronner-Fraser, M. (2003). Neural crest contributions tothe lamprey head. Development 130, 2317–2327.

McGuire, S.E., Le, P.T., Osborn, A.J., Matsumoto, K., and Davis, R.L. (2003).Spatiotemporal rescue of memory dysfunction in Drosophila. Science 302,1765–1768.

Meilhac, S.M., Esner, M., Kelly, R.G., Nicolas, J.F., and Buckingham, M.E.(2004). The clonal origin of myocardial cells in different regions of the embry-onic mouse heart. Dev. Cell 6, 685–698.

Meilhac, S.M., Adams, R.J., Morris, S.A., Danckaert, A., Le Garrec, J.F.,and Zernicka-Goetz, M. (2009). Active cell movements coupled to positionalinduction are involved in lineage segregation in the mouse blastocyst. Dev.Biol. 331, 210–221.

Mintz, B. (1965). Genetic Mosaicism in Adult Mice of Quadriparental Lineage.Science 148, 1232–1233.

Moody, S.A. (1987). Fates of the blastomeres of the 32-cell-stage Xenopusembryo. Dev. Biol. 122, 300–319.

Morata, G., and Ripoll, P. (1975). Minutes: mutants of drosophila autono-mously affecting cell division rate. Dev. Biol. 42, 211–221.

Mosimann, C., Kaufman, C.K., Li, P., Pugach, E.K., Tamplin, O.J., and Zon, L.I.(2011). Ubiquitous transgene expression and Cre-based recombination drivenby the ubiquitin promoter in zebrafish. Development 138, 169–177.

Muzumdar, M.D., Tasic, B., Miyamichi, K., Li, L., and Luo, L. (2007). A globaldouble-fluorescent Cre reporter mouse. Genesis 45, 593–605.

Naiche, L.A., and Papaioannou, V.E. (2007). Cre activity causes widespreadapoptosis and lethal anemia during embryonic development. Genesis 45,768–775.

Nicolas, J.F., Mathis, L., Bonnerot, C., and Saurin, W. (1996). Evidence inthe mouse for self-renewing stem cells in the formation of a segmentedlongitudinal structure, the myotome. Development 122, 2933–2946.

Nishida, H. (1987). Cell lineage analysis in ascidian embryos by intracellularinjection of a tracer enzyme. III. Up to the tissue restricted stage. Dev. Biol.121, 526–541.

Noctor, S.C., Flint, A.C., Weissman, T.A., Dammerman, R.S., and Kriegstein,A.R. (2001). Neurons derived from radial glial cells establish radial units inneocortex. Nature 409, 714–720.

Nowotschin, S., and Hadjantonakis, A.K. (2009). Use of KikGR a photoconver-tible green-to-red fluorescent protein for cell labeling and lineage analysis inES cells and mouse embryos. BMC Dev. Biol. 9, 49.

Nowotschin, S., Eakin, G.S., and Hadjantonakis, A.K. (2009). Live-imagingfluorescent proteins in mouse embryos: multi-dimensional, multi-spectralperspectives. Trends Biotechnol. 27, 266–276.

Olivier, N., Luengo-Oroz, M.A., Duloquin, L., Faure, E., Savy, T., Veilleux, I.,Solinas, X., Debarre, D., Bourgine, P., Santos, A., et al. (2010). Cell lineagereconstruction of early zebrafish embryos using label-free nonlinear micros-copy. Science 329, 967–971.

Perdigoto, C.N., Schweisguth, F., and Bardin, A.J. (2011). Distinct levels ofNotch activity for commitment and terminal differentiation of stem cells inthe adult fly intestine. Development, in press. 10.242/dev.065292.

Perea-Gomez, A., Lawson, K.A., Rhinn,M., Zakin, L., Brulet, P., Mazan, S., andAng, S.L. (2001). Otx2 is required for visceral endoderm movement and forthe restriction of posterior signals in the epiblast of the mouse embryo.Development 128, 753–765.

Peter, I.S., and Davidson, E.H. (2010). The endoderm gene regulatory networkin sea urchin embryos up to mid-blastula stage. Dev. Biol. 340, 188–199.

velopmental Cell 21, September 13, 2011 ª2011 Elsevier Inc. 407

Page 15: Developmental Cell Perspective - COnnecting REpositories · Developmental Cell Perspective Tracing Cells for Tracking Cell Lineage and Clonal Behavior Margaret E. Buckingham1, *and

Developmental Cell

Perspective

Petit, A.C., Legue, E., and Nicolas, J.F. (2005). Methods in clonal analysis andapplications. Reprod. Nutr. Dev. 45, 321–339.

Piatkevich, K.D., Hulit, J., Subach, O.M., Wu, B., Abdulla, A., Segall, J.E., andVerkhusha, V.V. (2010). Monomeric red fluorescent proteins with a largeStokes shift. Proc. Natl. Acad. Sci. USA 107, 5369–5374.

Plachta, N., Bollenbach, T., Pease, S., Fraser, S.E., and Pantazis, P. (2011).Oct4 kinetics predict cell lineage patterning in the early mammalian embryo.Nat. Cell Biol. 13, 117–123.

Placinta, M., Shen, M.C., Achermann, M., and Karlstrom, R.O. (2009). A laserpointer driven microheater for precise local heating and conditional generegulation in vivo. Microheater driven gene regulation in zebrafish. BMC Dev.Biol. 9, 73.

Plusa, B., Piliszek, A., Frankenberg, S., Artus, J., and Hadjantonakis, A.K.(2008). Distinct sequential cell behaviours direct primitive endoderm formationin the mouse blastocyst. Development 135, 3081–3091.

Reddy, G.V., Heisler, M.G., Ehrhardt, D.W., and Meyerowitz, E.M. (2004).Real-time lineage analysis reveals oriented cell divisions associated withmorphogenesis at the shoot apex of Arabidopsis thaliana. Development 131,4225–4237.

Rolland-Lagan, A.G., Coen, E., Impey, S.J., and Bangham, J.A. (2005). Acomputational method for inferring growth parameters and shape changesduring development based on clonal analysis. J. Theor. Biol. 232, 157–177.

Rosenquist, G.C., andDeHaan, R.L. (1966). Migration of precardiac cells in thechick embryo: a radioautographic study. Contrib. Embryol. 263, 111–121.

Rothenaigner, I., Krecsmarik, M., Hayes, J.A., Bahn, B., Lepier, A., Fortin, G.,Gotz, M., Jagasia, R., and Bally-Cuif, L. (2011). Clonal analysis by distinct viralvectors identifies bona fide neural stem cells in the adult zebrafish telenceph-alon and characterizes their division properties and fate. Development 138,1459–1469.

Sanes, J.R., Rubenstein, J.L., and Nicolas, J.F. (1986). Use of a recombinantretrovirus to study post-implantation cell lineage in mouse embryos. EMBOJ. 5, 3133–3142.

Sato, Y., Poynter, G., Huss, D., Filla, M.B., Czirok, A., Rongish, B.J., Little,C.D., Fraser, S.E., and Lansford, R. (2010). Dynamic analysis of vascularmorphogenesis using transgenic quail embryos. PLoS ONE 5, e12674.

Satoh, A., Sakamaki, K., Ide, H., and Tamura, K. (2005). Characteristics ofinitiation and early events for muscle development in the Xenopus limb bud.Dev. Dyn. 234, 846–857.

Schleiden, M.J. (1838). Beitrage zur Phytogenesis. In Muller’s Archiv furAnatomie, Physiologie und wissenschaftliche Medicin (Berlin: von Veit),pp. 137–176.

Schnabel, R., Hutter, H., Moerman, D., and Schnabel, H. (1997). Assessingnormal embryogenesis in Caenorhabditis elegans using a 4D microscope:variability of development and regional specification. Dev. Biol. 184, 234–265.

Schwann, T. (1839). Mikroskopische Untersuchungen uber die Ubereinstim-mung in der Struktur und dem Wachstume der Tiere und Pflanzen, G.E.Reimer, ed. (Berlin).

Serbedzija, G.N., Bronner-Fraser, M., and Fraser, S.E. (1989). A vital dye anal-ysis of the timing and pathways of avian trunk neural crest cell migration.Development 106, 809–816.

Shimomura, O., Johnson, F.H., and Saiga, Y. (1962). Extraction, purificationand properties of aequorin, a bioluminescent protein from the luminous hydro-medusan, Aequorea. J. Cell. Comp. Physiol. 59, 223–239.

Sinha, D.K., Neveu, P., Gagey, N., Aujard, I., Le Saux, T., Rampon, C., Gauron,C., Kawakami, K., Leucht, C., Bally-Cuif, L., et al. (2010). Photoactivation of theCreER T2 recombinase for conditional site-specific recombination with highspatiotemporal resolution. Zebrafish 7, 199–204.

Snippert, H.J., van der Flier, L.G., Sato, T., van Es, J.H., van den Born, M.,Kroon-Veenboer, C., Barker, N., Klein, A.M., van Rheenen, J., Simons, B.D.,and Clevers, H. (2010). Intestinal crypt homeostasis results from neutralcompetition between symmetrically dividing Lgr5 stem cells. Cell 143,134–144.

408 Developmental Cell 21, September 13, 2011 ª2011 Elsevier Inc.

Spemann, H., and Mangold, H. (1924). Uber Induktion von Embryonalanlagendurch Implantation artfremder Organisatoren. Wilhelm Roux Arch. Entw.Mech. Org. 100, 599–638.

Stark, D.A., and Kulesa, P.M. (2007). An in vivo comparison of photoactivat-able fluorescent proteins in an avian embryo model. Dev. Dyn. 236, 1583–1594.

Stent, G.S. (1985). The role of cell lineage in development. Philos. Trans. R.Soc. Lond. B Biol. Sci. 312, 3–19.

Stern, C. (1936). Somatic Crossing over and Segregation in DrosophilaMelanogaster. Genetics 21, 625–730.

Sturtevant, A.H. (1929). The claret mutant type of Drosophila simulans: Astudy of chromosome elimination and cel lineage. Z Wiss Zool Abt A 135,323–356.

Sulston, J.E., Schierenberg, E., White, J.G., and Thomson, J.N. (1983). Theembryonic cell lineage of the nematode Caenorhabditis elegans. Dev. Biol.100, 64–119.

Swoboda, P., Gal, S., Hohn, B., and Puchta, H. (1994). Intrachromosomalhomologous recombination in whole plants. EMBO J. 13, 484–489.

Swoger, J., Muzzopappa, M., Lopez-Schier, H., and Sharpe, J. (2011). 4Dretrospective lineage tracing using SPIM for zebrafish organogenesis studies.J Biophotonics 4, 122–134.

Tam, P.P., Parameswaran, M., Kinder, S.J., and Weinberger, R.P. (1997).The allocation of epiblast cells to the embryonic heart and other mesodermallineages: the role of ingression and tissue movement during gastrulation.Development 124, 1631–1642.

Turan, S., Galla, M., Ernst, E., Qiao, J., Voelkel, C., Schiedlmeier, B., Zehe,C., and Bode, J. (2011). Recombinase-mediated cassette exchange(RMCE): traditional concepts and current challenges. J. Mol. Biol. 407,193–221.

Tzouanacou, E., Wegener, A., Wymeersch, F.J., Wilson, V., and Nicolas, J.F.(2009). Redefining the progression of lineage segregations during mammalianembryogenesis by clonal analysis. Dev. Cell 17, 365–376.

Ueno, H., and Weissman, I.L. (2006). Clonal analysis of mouse developmentreveals a polyclonal origin for yolk sac blood islands. Dev. Cell 11, 519–533.

Vincent, J.P., and O’Farrell, P.H. (1992). The state of engrailed expressionis not clonally transmitted during early Drosophila development. Cell 68,923–931.

Vincent, C.A., Carpenter, R., and Coen, E.S. (1995). Cell lineage patterns andhomeotic gene activity during Antirrhinum flower development. Curr. Biol. 5,1449–1458.

Virchow, R. (1858). Die Cellularpathologie in ihrer Begrundung auf physiologi-sche und pathologische Gewebelehre, A. Hirschwald, ed. (Berlin).

Vogt, W. (1929). Gestaltungsanalyse am Amphibienkeim mit ortlicher Vitalfar-bung. II. Teil. Gastrulation une Mesodermbildung bei Anuren und Urodelen.Wilhelm Roux Arch. Entw. Mech. Org. 120, 384–706.

Voiculescu, O., Bertocchini, F., Wolpert, L., Keller, R.E., and Stern, C.D. (2007).The amniote primitive streak is defined by epithelial cell intercalation beforegastrulation. Nature 449, 1049–1052.

Waddington, C.H. (1932). Experiments on the development of chick and duckembryos. Philos. Trans. R. Soc. Lond. B Biol. Sci. 221, 179–230.

Walsh, C., and Cepko, C.L. (1992). Widespread dispersion of neuronal clonesacross functional regions of the cerebral cortex. Science 255, 434–440.

Wasserstrom, A., Adar, R., Shefer, G., Frumkin, D., Itzkovitz, S., Stern, T., Shur,I., Zangi, L., Kaplan, S., Harmelin, A., et al. (2008). Reconstruction of celllineage trees in mice. PLoS ONE 3, e1939.

Weisblat, D.A., Sawyer, R.T., and Stent, G.S. (1978). Cell lineage analysis byintracellular injection of a tracer enzyme. Science 202, 1295–1298.

Weston, J.A. (1963). A radioautographic analysis of the migration and localiza-tion of trunk neural crest cells in the chick. Dev. Biol. 6, 279–310.

Page 16: Developmental Cell Perspective - COnnecting REpositories · Developmental Cell Perspective Tracing Cells for Tracking Cell Lineage and Clonal Behavior Margaret E. Buckingham1, *and

Developmental Cell

Perspective

Wetzel, R. (1929). Untersuchungen am Huhnchen. Die Entwicklung des Keimswahrend der ersten beiden Bruttage. Wilhelm Roux Arch. Entw. Mech. Org.119, 188–321.

Whitman, C.O. (1887). A contribution to the history of germ layers in Clepsine.J. Morphol. 1, 105–182.

Wilson, E.B. (1892). The Cell-lineage of Nereis. A contribution to the cytogenyof the Annelid body. J. Morphol. VI, 368.

Xu, T., and Rubin, G.M. (1993). Analysis of genetic mosaics in developing andadult Drosophila tissues. Development 117, 1223–1237.

De

Yamanaka, S. (2009). A fresh look at iPS cells. Cell 137, 13–17.

Yu, H.H., Chen, C.H., Shi, L., Huang, Y., and Lee, T. (2009). Twin-spot MARCMto reveal the developmental origin and identity of neurons. Nat. Neurosci. 12,947–953.

Zinyk, D.L., Mercer, E.H., Harris, E., Anderson, D.J., and Joyner, A.L. (1998).Fate mapping of the mouse midbrain-hindbrain constriction using a site-specific recombination system. Curr. Biol. 8, 665–668.

Zong, H., Espinosa, J.S., Su, H.H., Muzumdar, M.D., and Luo, L. (2005).Mosaic analysis with double markers in mice. Cell 121, 479–492.

velopmental Cell 21, September 13, 2011 ª2011 Elsevier Inc. 409


Recommended