Download as pdf or txt
Download as pdf or txt
You are on page 1of 25

REVIEWS

THE BIOLOGICAL RESTORATION OF CENTRAL


NERVOUS SYSTEM ARCHITECTURE AND FUNCTION:
PART 1—FOUNDATIONS AND HISTORICAL
LANDMARKS IN CONTEMPORARY STEM CELL BIOLOGY
Azadeh Farin, M.D.
Department of Neurological Surgery, SINCE THEIR DISCOVERY, stem cells have fascinated scientists with their ultimate poten-
Keck School of Medicine, tial: the ability to cure disease, repair altered physiology, and reverse neurological deficit.
University of Southern California, Stem cell science unquestionably promises to eliminate many of the tragic limitations con-
Los Angeles, California
temporary medicine must acknowledge, and cloning may provide young cells for an aging
population. Although it is widely believed that stem cells will transform the way medicine
Charles Y. Liu, M.D., Ph.D.
is practiced, therapeutic interventions using stem cell technology are still in their infancy.
Department of Neurological Surgery,
Keck School of Medicine, The 3 most common stem cell sources studied today are umbilical cord blood, bone mar-
University of Southern California, row, and human embryos. Although cord blood is currently used to treat dozens of disor-
Los Angeles, California, and ders and bone marrow stem cells have been used clinically since the 1960s, human embry-
Division of Chemistry and
Chemical Engineering,
onic stem cells have yet to be successfully applied to any disease. Undeniably, stem cell
California Institute of Technology, therapy has the potential to be one of the most powerful therapeutic options available. In
Pasadena, California this introductory article of a 5-part series on stem cells, we narrate the evolution of modern
stem cell science, delineating major landmarks that will prove responsible for taking stem
James B. Elder, M.D. cell technology from the laboratory into revolutionary clinical applications: from the first mile-
Department of Neurological Surgery, stone of identifying the mouse hematopoietic stem cell to the latest feats of producing
Keck School of Medicine,
University of Southern California,
pluripotent stem cells without embryos at all. In Part 2, we present the evidence demon-
Los Angeles, California strating the certainty of adult mammalian neurogenesis; in Parts 3 and 4, we describe neu-
rosurgical applications of stem cell technology; and in Part 5, we discuss the philosophical
Iver A. Langmoen, M.D., Ph.D. and ethical issues surrounding stem cell therapy, as well as future areas of exploration.
Vilhelm Magnus Center,
KEY WORDS: Cellular neurosurgery, Cloning, Embryo, Molecular neurosurgery, Neurogenesis, Plasticity,
Institute for Surgical Research,
and Department of Neurosurgery,
Stem cell
Ullevål University Hospital and
Rikshospitalet, Neurosurgery 64:15–39, 2009 DOI: 10.1227/01.NEU.0000337580.02706.DC www.neurosurgery-online.com
University of Oslo, Norway, and
Department of Clinical Neuroscience,

T
Karolinska Institute, he discovery of stem cells is considered a testing, and understanding the earliest stages of
Stockholm, Sweden breakthrough that is expected to pro- human development are enormous. These
foundly impact the practice of medicine. notions are just now beginning to emerge as
Michael L.J. Apuzzo, M.D. Although most studies have been historically actualities. In this article, we recount the most
Department of Neurological Surgery, conducted in murine models, advances in critical feats in modern stem cell research that
Keck School of Medicine,
human embryonic stem (ES) cell research are will lead to definitive clinical applications. A
University of Southern California,
Los Angeles, California now a reality. Human ES cells are currently glossary is provided in the Appendix.
derived from 3 sources: blastocysts remaining
Reprint requests: after infertility treatments and donated for DEFINITION OF STEM CELLS
Azadeh Farin, M.D., research, blastocysts generated from donated
Department of Neurological Surgery, gametes, and nuclear transfer. The potential During the 1800s, the cell was identified as
Los Angeles County-University of
Southern California Medical Center,
implications for treating human disease, drug the building block of life. Mammals were then
1200 North State Street, No. 5046,
Los Angeles, CA 90033. ABBREVIATIONS: CBE, cord blood-derived embryonic-like stem cell; DNA, deoxyribonucleic acid; ES, embry-
Email: Azadehfarin@hotmail.com onic stem; HSC, hematopoietic stem cell; iPS, induced pluripotent stem; Oct4, Octamer 4; SCID, severe com-
bined immunodeficiency; SCNT, somatic cell nuclear transfer; SHED, stem cells from human exfoliated decidu-
Received, May 6, 2008.
ous teeth; Sox2, sex-determining region Y-box 2
Accepted, September 16, 2008.

NEUROSURGERY VOLUME 64 | NUMBER 1 | JANUARY 2009 | 15


FARIN ET AL.

determined to be composed of 3 cell types: germ cells, somatic oocyte and sperm. This totipotent stem cell, or “master cell,”
cells, and stem cells. Germ cells are the haploid gametes (oocyte contains all of the genetic information needed to become any tis-
and spermatocyte) responsible for reproductive capacity; oocyte sue whatsoever, including placenta (Fig. 1). Cells produced by
fertilization by a spermatocyte results in a diploid zygote with the first few divisions of the fertilized egg are also totipotent.
46 chromosomes. The majority of cells in the body are diploid The morula becomes the blastocyst, which, in humans, is a 4- to
somatic cells. The word “somatic” is derived from soma the 5-day-old embryo consisting of 50 to 150 cells. The blastocyst
Greek word for body. In the 1800s, the observation that certain then generates 3 main types of stem cells—ES cells, umbilical
cells could generate other cells marked the commencement of cord blood stem cells, and adult stem cells (Fig. 2)—as it
stem cell science. This was followed in the early 1900s by the matures into the human fetus.
discovery of the first hematopoietic stem cells (HSC) differen-
tiating into blood cells. Pluripotent ES Cells
Stem cells, by definition, are primary undifferentiated cells ES cells are pluripotent cells derived from either the blasto-
possessing 2 properties: self-renewal and potency. Self-renewal cyst’s undifferentiated inner cell mass epiblast tissue or from the
is illustrated by the retained ability of stem cells to renew them- morula. In contrast to multipotent adult stem cells, ES cells can
selves through endless mitotic cycles while maintaining the differentiate during development into all specialized embry-
undifferentiated state. To ensure self- renewal, stem cells onic tissues derived from the 3 primary germ layers (ectoderm,
undergo 2 types of cell division. Symmetric division gives rise endoderm, and mesoderm) (Fig. 1). They can further differenti-
to 2 identical daughter cells, both of which are endowed with ate into each of more than 200 cell types of the adult body when
stem cell properties. Asymmetric division produces only 1 stem given sufficient and necessary stimulation for a specific cell lin-
cell and a progenitor cell with limited self-renewal potential. eage (Fig. 3). Unlike the totipotent zygote, however, these cells
Progenitors can undergo several cycles of cell division before do not contribute to the extraembryonic membranes, placenta,
terminally differentiating into a mature cell. The molecular and some uterine tissues.
switch determining symmetric versus asymmetric divisions Most research has involved mouse ES cells and human ES
may be dependent on differential segregation of cell membrane cells. Differences between these cell types have been extensively
proteins, such as receptors, between daughter cells (6). described and are illustrated in Figures 4 and 5 (1, 3, 4, 7, 9, 10,
Alternatively, stem cells may remain undifferentiated owing to 12, 15, 17, 22–24, 30, 31, 33, 35, 37–40, 45–48, 52, 55–62, 70, 72, 73,
environmental cues in their particular niche and may differen- 75–78, 81, 87, 89, 97, 98, 100, 103, 104, 108, 109, 113, 115, 117, 118,
tiate when they exit that niche or are deprived of certain signals. 120, 123, 139, 142, 144). To maintain an undifferentiated state,
For example, studies in Drosophila germarium have identified both mouse ES cells and human ES cells require very specific
the signals Dpp and adherens junctions that prevent germarium environments; without optimal culture conditions or genetic
stem cells from differentiating (110, 141). manipulation (11a), ES cells will rapidly differentiate. Mouse
Potency is defined as the potential to differentiate into special- ES cells are grown on a layer of gelatin and require the presence
ized cell types (49, 51, 90–92, 94, 101, 106, 112, 114, 135, 137). of leukemia-inhibitory factor (16).
Totipotent or pluripotent stem cells, or ES cells, are able to give Human ES cells are grown on a feeder layer of mouse embry-
rise to any or many mature cell types. Multipotent or unipotent onic fibroblasts and require the presence of basic fibroblast
progenitor cells are sometimes referred to as adult stem cells, growth factor (or fibroblast growth factor 2) (79). Human ES
and they are more limited in the variety of progeny they pro- cells maintain pluripotency owing to the action of 3 hallmark
duce. Considerable debate exists about whether some proposed transcription factors, NANOG, Octamer 4 (Oct4), and Sex-
adult cell populations are truly stem cells. determining region Y-box 2 (Sox2), which suppress genes lead-
Self-renewal and potency are illustrated through multiple ing to differentiation (8). These factors are the subjects of intense
cycles of regeneration in vivo. The “gold standard” test for defin- investigation aiming to uncode the mechanisms responsible for
ing an HSC is the ability to transplant an HSC into an individual cell pluripotency and to determine how these events can be
without HSCs and to rescue that individual; HSCs isolated from manipulated for human benefit. Thus far, all 3 factors are known
the individual who received the new transplant can subse- to play essential roles in the self-renewal of undifferentiated cells
quently be transplanted into another individual without HSCs, and maintenance of pluripotency. They also facilitate the tran-
demonstrating self-renewal. HSCs must demonstrate potency scription of developmentally important homeodomain proteins.
by producing new blood and immune cells. Stem cell properties The murine NANOG gene was isolated by Ian Chambers, who
can also be illustrated in vitro using clonogenic assays, in which summarized the significance of this gene by calling it the “mas-
single cells are characterized by their ability to differentiate and ter gene . . . [making ES cells] immortal” (11, 11a). NANOG
self-renew (26, 27). However, in vitro culture conditions can alter overexpression causes mouse ES cells to self-renew in the
cell behavior, rendering it difficult to assess whether the cells will absence of leukemia-inhibitory factor and human ES cells to
behave in a similar manner in vivo. propagate for multiple passages as pluripotent cells; gene knock-
Stem cells’ potential for unlimited versus limited differentia- down of NANOG and Oct4 promotes differentiation. Tumor
tion, or phenotypic maturation along a restricted exclusive lin- suppressor p53 suppresses NANOG expression after deoxyri-
eage, is a fundamental feature that can be used to categorize bonucleic acid (DNA) damage in mouse ES cells, inducing differ-
them. A morula, or zygote, is the product of fusion between entiation of cells, followed by cell cycle arrest and apoptosis.

16 | VOLUME 64 | NUMBER 1 | JANUARY 2009 www.neurosurgery-online.com


FOUNDATIONS AND LANDMARKS IN STEM CELL BIOLOGY

specific signals for correct dif-


ferentiation; if injected directly
into the body, ES cells differen-
tiate into many different types
of cells, resulting in a teratoma.
Differentiating ES cells for
practical purposes while avoid-
ing transplant rejection are 2
of the many technical issues
researchers face (140).
Significant ethical contro-
versy has been generated over
human ES cell research. It is
fueled by the fact that starting
an ES cell line has traditionally
required the destruction of a
human embryo or therapeutic
cloning. These techniques are
felt by some to devalue human
life (34, 132). Although ES cells
are considered to have greater
therapeutic potential than adult
stem cells, lack of consensus
with regard to associated com-
plex ethical concerns has prom-
pted some governments to re-
strict state-sponsored ES cell
activities. Significant ES cell re-
search has continued fruitfully
with private funds. However,
given the challenging curative
goals proposed for these cells,
further developments are re-
FIGURE 1. Development and differentiation of cells originating from the zygote (from, National Center for quired before ES cells can be
Biotechnology Information. http://www.ncbi.nlm.nih.gov. Accessed May 1, 2008 [76]). applied safely and effectively to
human disease. Consequently,
Oct4 is active in embryos throughout the preimplantation period after 20 years of research, there have been no approved treat-
and is a marker for both undifferentiated cells and tumors. Its ments or human trials using ES cells. Researchers have begun
exact level determines ES cell fate and regulates pluripotency. exploring approaches that obviate embryo destruction (Fig. 6).
Mouse embryos deficient in Oct4 fail to form the inner cell mass, Recently, it has been shown that ES cell lines can be generated
lose pluripotency, and differentiate into trophectoderm. An using a single-cell biopsy similar to that used in preimplantation
increase in Oct4 causes differentiation into primitive endoderm genetic diagnosis. The differentiated cell is reprogrammed to a
and mesoderm. Because stem cell self-renewal is dependent pluripotent state, and embryos are avoided altogether (83, 122).
upon a critical amount of Oct4 and deviations result in abnormal The ethical issues surrounding ES research will be further dis-
development, Oct4 is considered a primary regulator of pluripo- cussed in Part 5 of this series.
tency. The role of Oct4 in sustaining self-renewal capacity of
adult somatic stem cells requires further study. Sox2 is also Multipotent Adult Stem Cells
involved in regulation of embryonic development and cell fate Adult stem cells are differentiated from pluripotent inner mass
determination. cells. Most adult stem cells are lineage-restricted (multipotent),
The cell surface antigens most commonly used to identify only producing closely related progeny of a given local tissue
human ES cells are the glycolipids SSEA3 and SSEA4 and the ker- source (e.g., HSCs differentiate into red blood cells, white blood
atan sulfate antigens Tra-1-60 and Tra-1-81. The molecular defini- cells, and platelets). These undifferentiated cells reside with dif-
tion of a stem cell includes many more proteins and continues to ferentiated cells of their tissues of origin, with correlative nomen-
be a topic of research (40). Because of their potential for unlimited clature (mesenchymal stem cell, adipose-derived stem cell,
expansion and pluripotency, ES cells remain a theoretically attrac- endothelial stem cell) (5, 32). Adult stem and progenitor cells
tive source for regenerative tissue. However, ES cells require play a reparative role, replenishing cells and maintaining normal

NEUROSURGERY VOLUME 64 | NUMBER 1 | JANUARY 2009 | 17


FARIN ET AL.

EVOLUTION
OF MODERN
STEM CELL SCIENCE
1963: Discovery of
the Mouse HSC
The first major discovery in
modern stem cell science is
attributed to Canadian scien-
tists Ernest A. McCulloch and
James E. Till, whose decade-
long experiments culminated
in the 1963 discovery of the
multipotent HSC in mouse
bone marrow (129). The HSC
was actually isolated years
later. Contributing to this dis-
covery was knowledge that
radiation from nuclear bombs
and x-rays can eliminate blood
cell production and that blood
FIGURE 2. Classification of human stem cells (from, Bongso A, Lee EH: Stem cells: Their definition, classification
and sources, in Bongso A, Lee EH (eds): Stem Cells: From Bench to Bedside. Singapore, World Scientific, cell production could be res-
2005, pp 1–13 [7], at http://www.worldscibooks.com/lifesci/etextbook/5729/5729_chap1.pdf. Accessed May 1, 2008). cued by bone marrow injec-
tions from healthy mice.
Starting in the 1950s, Mc-
Cullough and Till systemati-
turnover of regenerative organs, such as blood, skin, or intestinal cally measured the quantity of donor bone marrow cells needed
tissues. Given their functions, they are also known as somatic to restore blood cell production in irradiated mice. These meas-
stem cells or germline stem cells, as they give rise to gametes. urements enabled them to deduce that hematopoietic rescue
Despite their name, they can be found in children as well (41). was being carried out by a very small number of donor bone
A major research focus has been to clarify their capacity for marrow cells. They further observed that the spleens of the irra-
indefinite self-renewal and to better characterize their differen- diated mice receiving replacement bone marrow contained
tiation potential (28). Pluripotent adult stem cells are rare and colonies of red blood cells, white blood cells, and platelets, indi-
generally small in number, but they can be found in a number cating that something capable of making all blood cell types, a
of tissues, including umbilical cord blood (69, 99). blood-forming stem cell, resided in the spleen. In a truly elegant
Because the production of adult stem cells does not involve experiment, McCullough and Till imparted distinct genetic sig-
embryo destruction, related research attracts greater govern- natures to blood-forming stem cells in donor bone marrow and
ment funding, and therapeutic applications have been wide- documented that the blood cells in each spleen colony bore a
spread (133). Whereas ES cell potential remains untested, adult particular genetic imprint matching that of the donor stem cell.
stem cell treatments have been used for many years to success- This proved that diverse blood cells come from an individual
fully treat leukemia and related bone and blood cancers stem cell. McCullough and Till demonstrated this self-renewal
through bone marrow transplants. Promising investigations potential of stem cells by repeatedly taking individual spleen
involve antidifferentiation transcription factors (oncogene c- colonies from a set of mice and injecting them into irradiated
Myc) that signal reprogramming pathways to transform adult mice to generate new colonies. Their contributions, which initi-
stem cells to an embryonic-like state, reversing differentiation ated the efforts to define how, and in response to what, stem
and recapacitating adult cells with pluripotency (121). How- cells divide and mature, serve as a foundation for all current
ever, oncogenic transformation may limit the clinical appro- research on stem cells (65–68, 80, 93, 126–131).
priateness of this technique.
1968: Bone Marrow Transplant Successfully Treats
Unipotent Stem Cells Human Severe Combined Immunodeficiency
Unipotent cells give rise to only 1 cell type but have the prop- The next major milestone in stem cell science occurred in 1968
erty of self-renewal, which distinguishes them from non-stem at the University of Minnesota, when severe combined immun-
cells. Some researchers claim unipotency to be inadequate in odeficiency (SCID) was successfully treated by the first success-
terms of defining a stem cell, arguing that at least multipotency ful bone marrow transplant between 2 human siblings (21, 29).
is required. Examples of unipotent cells are human skin cells. This primary immune deficiency is characterized by a severe

18 | VOLUME 64 | NUMBER 1 | JANUARY 2009 www.neurosurgery-online.com


FOUNDATIONS AND LANDMARKS IN STEM CELL BIOLOGY

FIGURE 3. In vitro avenues to differentiation of embryonic stem cells. tor; EB, embryoid body; IGF2, insulin-like growth factor 2; NTF3, neu-
Several in vitro embryonic stem cell differentiation protocols show how a rotrophin 3; SDIA, stromal-derived inducing activity method; SHH, sonic
defined microenvironment can enhance various differentiation pathways by hedgehog homologue; VEGF, vascular endothelial growth factor (from,
partially imitating early embryonic development. bFGF, basic fibroblast Klimanskaya I, Rosenthal N, Lanza R: Derive and conquer: Sourcing and
growth factor (also known as fibroblast growth factor 2); BMP2/4, bone differentiating stem cells for therapeutic applications. Nat Rev Drug
morphogenetic protein 2/4; dHGF, deleted variant of hepatocyte growth fac- Discov 7:131–142, 2008 [47]).

defect in the T- and B-lymphocyte systems caused by absent or whereas SCID is usually fatal within 1 year after birth without
poorly functioning lymphocytes or defective bone marrow stem transplantation. Unfortunately, more than 90% of patients with
cells, from which mature lymphocytes normally develop. SCID primary immunodeficiencies lack suitably histocompatible sib-
renders its infant victims prone to life-threatening pneumonia, ling donors. Alternative sources of stem cells are being investi-
meningitis, or sepsis within the first few months of life; even gated to treat these patients. Bone marrow from closely matched
normal childhood infections and viruses (chicken pox, measles, relatives and closely matched unrelated individuals, peripheral
and cold sore virus) may be fatal. The term “bubble baby” has blood stem cells, and cord blood from siblings or unrelated new-
been used to describe such babies because of the sterile environ- borns have all been used successfully in selected cases.
ment used to decrease the risk of infections.
Marrow transplantation is the only treatment that has been 1978: HSCs Discovered in Human Cord Blood
shown to cure SCID and other prematurely lethal primary The treatment of blood disorders was advanced further by
immunodeficiencies. A 90% cure rate can be expected for SCID, the discovery of stem cells in cord blood (95). Rich in HSCs, cord

NEUROSURGERY VOLUME 64 | NUMBER 1 | JANUARY 2009 | 19


FARIN ET AL.

Gail R. Martin were the first


workers to isolate mouse pluri-
potent ES cells from mouse
embryos and condition them
to differentiate into teratocarci-
nomas when injected in vivo.
Cultures of these ES cells dif-
ferentiated into a wide variety
of cell types, making the full
potential of ES cells a reality.
No longer did it appear that
therapy would be limited by
the multipotency of the HSC
instead of the more desirable
pluripotency of ES cells. Gail
Martin is credited with coining
the term “embryonic stem cell”
(64). In her article, she refers to
permissive culture conditions
that promote stem cell proper-
ties, and she further describes
the implications for patho-
physiological discoveries re-
lated to her technique: “. . .
Embryonic stem cells were iso-
lated from . . . blastocysts cul-
tured in medium . . . [which]
might contain a growth factor
that stimulates the proliferation
or inhibits the differentiation of
. . . embryonic cells. . . . This
method . . . makes feasible the
FIGURE 4. Comparison of mouse, monkey, and human pluripotent stem cells. ES cell, embryonic stem cell; EG, isolation of pluripotent cell lines
embryonic germ cell; EC, embryonic carcinoma cell; SSEA, stage-specific embryonic antigen; TRA-1, tumor rejec-
from . . . embryo . . . carrying
tion antigen-1; LIF, leukemia-inhibitory factor; bFGF, basic fibroblast growth factor; EB, embryoid bodies; gp, glyco-
protein (adapted from, NIH Stem Cell Information. http://stemcells.nih.gov. Accessed May 1, 2008 [79]).
mutant genes . . . [and] should
make possible new approaches
to the study of early mam-
blood is the blood remaining in the placenta and umbilical cord, malian development” (64, p 7634).
which can be accessed during birth. Since 1988, scientists have Two years later, Elizabeth J. Robertson demonstrated that stem
used cord blood stem cells to treat leukemia, thalassemia, and cells isolated from parthenogenetic mouse embryos could form
anaplastic anemia. The unique genetic signature of cord blood a variety of tissues, including nerve and muscle (44, 102).
stem cells is partly responsible for higher success rates in patients
who receive a transplant of cord blood stem cells from a family 1992: Rat Neural Stem Cells Cultured
member as compared with cells from a nonrelative or even bone in Vitro as Neurospheres
marrow-derived HSCs. Cord blood collection is relatively easy Investigators at the California Institute of Technology first
and free of risk to the donor. Families with histories of hemato- isolated multipotent neural stem cells from the mammalian neu-
logical disorders are encouraged to consider banking their baby’s ral crest in 1992. These neural stem cells generated both multi-
umbilical cord blood for potential future therapy. It is vital that potent progeny, illustrating the self-renewal definition of stem
cord blood stem cells are collected within 15 minutes after birth cells, as well as committed clonal progeny that differentiated
and processed within 48 minutes at the cord blood bank’s labo- into neurons and glia (116). Progeny fate depended on substrate
ratory to preserve cell viability. manipulation. The authors wrote, “These findings have implica-
tions for models of neural crest development in vivo, and estab-
1981: Mouse ES Cells Derived from the Inner Cell Mass lish a system for studying the generation of cellular diversity by
Before 1981, HSCs were a common research focus, whereas a multipotent stem cell in vitro” (116, p 973).
the potential of ES cells was merely theoretical. Then, in a land- Concurrently, cloning technology was being developed at an
mark article, scientists Martin Evans, Matthew Kaufman, and alarming rate, far faster than most of the lay public could com-

20 | VOLUME 64 | NUMBER 1 | JANUARY 2009 www.neurosurgery-online.com


FOUNDATIONS AND LANDMARKS IN STEM CELL BIOLOGY

specific cell surface markers for


undifferentiated cells that were
distinct from murine markers.
The latter observation implies
that basic species differences
exist between early mouse and
human development. Even
after months of undifferenti-
ated in vitro proliferation, the
cells maintained pluripotency.
The cell line is still in use today.
Thomson aptly wrote, “Em-
bryonic stem cells provide a
powerful approach for intro-
ducing specific genetic chang-
es” (125, p 1145), a fact that has
since figured prominently in
biomedical research through
the generation of knockout
mice and today can be ack-
nowledged as a feature of para-
mount benefit to mankind. In
his article, Thomson elucidates
the potential utility of his find-
ing in advancing mammalian
FIGURE 5. Comparison of mouse and human embryonic stem cells. SSEA, stage-specific embryonic antigen; TRA-1, developmental biology and
tumor rejection antigen-1; N/A, not applicable (adapted from, Klimanskaya I, Rosenthal N, Lanza R: Derive and con- drug discovery: “Screens based
quer: Sourcing and differentiating stem cells for therapeutic applications. Nat Rev Drug Discov 7:131–142, 2008 [47]).
on the in vitro differentiation of
human ES cells to specific line-
prehend. Achievements in stem cell science for the next few ages could identify gene targets for new drugs, genes that could
years alternated between impressive technical feats credited to be used for tissue regeneration therapies, and teratogenic or toxic
those advancing the field of cloning and those working with compounds” (125, p 1146). He correctly predicted particular ben-
pluripotent stem cells. efit with regard to developmental events that cannot be studied
directly in the intact human embryo, including birth defects,
1996: First Mammal Cloned from Adult Somatic Cell infertility, pregnancy loss, and early postimplantation develop-
In 1996, the ewe Dolly was the first mammal to be cloned ment. Thomson foresaw the value of human ES cells in studying
from an adult somatic cell, using the process of nuclear transfer the development and function of tissues that differ between mice
(53, 54, 105, 138), which will be elaborated on later. She was and humans, including placenta and extraembryonic mem-
cloned by Ian Wilmut, Keith Campbell, and colleagues at the branes. Finally, he alluded to future aims in transplantation med-
Roslin Institute in Edinburgh, Scotland. This pioneering research icine, articulating that clarifying the mechanisms controlling dif-
helped create the basis for therapeutic and reproductive cloning. ferentiation was a prerequisite to the proficient, directed
Dolly lived until the age of 6 years. She was documented to have differentiation of ES cells to specialized types of functional cells
telomeres almost the same length as her mother’s, much shorter
with physiological relevance. He outlined that the consistent
than those of other sheep that were physiologically her age. Most
manufacturing of large quantities of human cells such as neu-
sheep live until about 13 years. Being cloned from an older sheep
rons would potentially permit cell replacement capacities in neu-
was implicated as a potential cause for her shortened telomeres
rodegenerative diseases. Challenges included “. . . strategies to
and early death; others think that her autopsy-proven pul-
prevent immune rejection of the transplanted cells . . . [these]
monary disease explains her early death.
include banking ES cells with defined major histocompatibility
1998: First Human ES Cell Line Derived complex backgrounds or genetically manipulating ES cells to . . .
In 1998, James Thomson derived the first human pluripotent combat immune rejection. . . . Substantial advances in basic
ES cell line at the University of Wisconsin-Madison (125) from developmental biology are required to direct ES cells efficiently
human blastocysts, 18 years after Gail Martin isolated mouse ES to lineages of human clinical importance” (125, p 1147).
cells. This landmark feat is widely cited as one of the most crit-
ical breakthroughs in modern stem cell science. 2001: Scientists Clone First Human Embryos
The human ES cells exhibited normal karyotypes, formed deriv- On October 31, 2001, 4 years after Dolly was cloned, scientists at
ative tissues of all 3 embryonic layers, and expressed primate- Advanced Cell Technology in Worcester, MA, cloned the first 4- to

NEUROSURGERY VOLUME 64 | NUMBER 1 | JANUARY 2009 | 21


FARIN ET AL.

Women between the ages of 24


A to 32 who had at least 1 child
and were willing to contribute
eggs anonymously were re-
cruited while skin biopsies
were collected from individu-
als to be cloned (the donors).
The skin biopsies were used to
isolate fibroblasts, and their
donors included patients with
diabetes or spinal cord injury
B because of the potential benefit
to them from therapeutic
cloning.
The technique is described
and illustrated in basic terms
(Fig. 7). First, an extremely fine
needle was used to aspirate the
C genetic material from a mature
egg; the donor cell nucleus or
entire cell was then injected
into the enucleated egg. The
transplanted egg was incubated
under conditions prompting
FIGURE 6. Avenues to generating pluripotent cell derivatives. A, somatic cell nuclear transfer (SCNT). The nucleus division and growth. The tim-
of the fertilized oocyte is removed, and the nucleus of a recipient’s somatic cell is transferred into the enucleated oocyte. ing of each cloning attempt
The somatic nucleus is reprogrammed by the competent cytoplasm, and the oocyte with the donor nucleus starts cleav- was dependent upon the men-
ing and forms a morula that continues to develop into a blastocyst. Pluripotent embryonic stem cells (ESC) with the strual cycles of the egg contrib-
recipient’s genome are derived from either the morula or the inner cell mass of the blastocyst. Their differentiated deriv- utors, who were injected with
atives are immunologically identical to the recipient’s own. B, parthenote pluripotent cells. An unfertilized oocyte is hormones so that they would
activated and starts to cleave as a normal embryo. Pluripotent cells are derived from a morula or the inner cell mass ovulate many eggs at once.
of a blastocyst. The absence of the paternal contribution greatly diminishes the human leukocyte antigen (HLA) vari- Initial failure led to adoption of
ability and thus allows a bank of various HLA types of pluripotent cells and/or their derivatives to be built. C, trans-
a modified technique, devel-
differentiation. The recipient’s somatic cell is turned into a pluripotent cell via cellular reprogramming using trans-
oped by Teruhiko Wakayama,
fection of a defined set of transcription factors or loading the target cell with the cellular lysate of a pluripotent cell.
The new pluripotent cell and its derivatives are immunologically identical to the recipient’s and so can be used for who created the first cloned
transplantation (from, Klimanskaya I, Rosenthal N, Lanza R: Derive and conquer: Sourcing and differentiating stem mice in 1998. Some eggs were
cells for therapeutic applications. Nat Rev Drug Discov 7:131–142, 2008 [47]). injected with nuclei from skin
fibroblasts, while others were
injected with ovarian cells
6-cell human embryos (14, 53, 54). The nuclear transplantation called cumulus cells that usually nurture developing eggs in the
experiments were reportedly conducted to develop blastocysts to ovary and can be found still attached to eggs after ovulation.
isolate human ES cells and develop replacement cells and tissues Cumulus cells are small enough to be injected whole. Ultimately,
for therapeutic purposes. Only 1 embryo progressed to the 6-cell 71 eggs from 7 volunteers were required before the first cloned
stage, at which point it stopped dividing (the blastocyst stage embryo was generated.
would have required 50–100 cells). The scientists did successfully The scientists then turned their attention to parthenogenesis
develop blastocysts parthenogenetically by prompting human eggs (asexual reproduction), whereby they induced human eggs to
alone, without sperm or other additional nuclear material, to divide into early embryos without being fertilized by a sperm or
repeatedly divide. The authors noted that their accomplishment without being enucleated and injected with a donor cell. Because
“[represented] the dawn of a new age in medicine by demonstrat- eggs proceed from the diploid state to the haploid state late in
ing that the goal of therapeutic cloning is within reach” (15, p 44). their maturation cycle, they can be isolated while they still retain
They defended their objective of therapeutic cloning as distinct a full set of genes. This technique was felt to be advantageous
from reproductive cloning, a practice to which they are opposed. because stem cells derived from such parthenogenetically acti-
Whereas therapeutic cloning attempts to use a patient’s own vated cells would be unlikely to be rejected after transplantation
genetic material to generate cells or tissue to treat disease or dys- because of their extreme similarity to a patient’s own cells. On the
function, reproductive cloning seeks to implant a cloned embryo other hand, they would not be identical to the individual’s cells
into a woman’s uterus with hopes of developing a cloned baby. because of the obligatory gene shuffling that occurs during ooge-

22 | VOLUME 64 | NUMBER 1 | JANUARY 2009 www.neurosurgery-online.com


FOUNDATIONS AND LANDMARKS IN STEM CELL BIOLOGY

FIGURE 7. Therapeutic cloning (from, Cibelli JB, Lanza RP, West Scientific American Online. http://cmbi.bjmu.edu.cn/news/0111/76.htm.
MD, Ezzell C: The first human cloned embryo. Sci Am 286:44–51, 2002, at Accessed May 1, 2008 [15]).

nesis. Cells derived parthenogenetically might also be more extracted and cultured the pulp, isolated viable multipotent
acceptable to those who are opposed to derivation of stem cells stem cells, and discovered that about 12 to 20 stem cells from
from cloned early embryos. The scientists described a scenario in each tooth could reliably grow in culture. The cells were named
which a woman with heart disease has her own eggs collected stem cells from human exfoliated deciduous teeth (SHED).
and activated in the laboratory to yield blastocysts. Stem cells iso- Children normally develop a set of 20 deciduous teeth, which
lated from the blastocyst could be chemically induced to become appear after 6 months of life and are replaced between ages 6
cardiac muscle cells, which are then implanted back into the and 12. These relatively accessible adult multipotent stem cells
patient to repair the diseased heart. Androgenesis, in which a were found to be highly proliferative; compared with dental
zygote containing only paternal chromosomes is developed, pulp stem cells from permanent teeth, SHED divide much more
could conceivably create autologous stem cells to treat a man. The rapidly, suggesting that SHED may be in a more immature state
scientists exposed 22 eggs to a chemical environment that than adult stem cells. Furthermore, these clonogenic cells could
changed the concentration of intracellular ions. After 5 days of be induced to express surface proteins, reflecting their transi-
growing in culture, 6 eggs had developed into what appeared to tion from stem cells to neural cells, adipocytes, and odontoblasts.
be blastocysts, but none clearly contained the inner cell mass After in vivo transplantation, SHED could induce bone forma-
required to isolate stem cells. tion, generate dentin, and survive in mouse brain while express-
The next few years witnessed astounding technical feats with ing neural markers (Fig. 8). Deciduous teeth are believed to be
the aim of deriving multipotent, and preferably pluripotent, capable of providing enough stem cells for potential clinical
stem cells without requiring embryos, given the controversy application, including autologous stem cell transplantation to
generated by the traditional technique of deriving pluripotent aid in the repair of damaged teeth, regenerate bone, and treat
stem cells at the cost of embryonal destruction. Stem cells were neurological diseases. Future efforts involve comprehensively
discovered in dental pulp, umbilical cord blood, and amniotic detailing all the cell types that can be generated from SHED.
fluid. Eventually, pluripotent stem cells were derived in mice
without embryos at all, and, subsequently, human hepatocytes 2005: Human Umbilical Cord Blood-derived Embryonic-
were generated from umbilical cord stem cells, not ES cells. Not like Stem Cells Discovered
surprisingly, human induced pluripotent stem (iPS) cells soon Researchers at Kingston University in England claimed to have
followed, and subsequently, human ES cell lines were generated discovered a third category of stem cells, cord blood-derived
without embryonal destruction. embryonic-like stem cells (CBE). The group claimed that these
cells are more potent than adult stem cells (69). After elective
2003: Multipotent Adult Stem/Postnatal Cells Caesarean section, umbilical cord blood components are sepa-
Discovered in Children’s Primary Teeth rated immunomagnetically. Cells are then cultured, and CBE
Dr. Songtao Shi of the National Institutes of Health’s National colonies are isolated. These colonies exhibit the human stem cell
Institute of Dental and Craniofacial Research (74, 107) discov- restricted markers TRA-1-60, TRA-1-81, SSEA-4, SSEA-3, and
ered a unique and rich supply of adult stem or postnatal cells in Oct4. CBEs cultured with hepatocyte growth medium resulted in
a naturally exfoliated human organ, the dental pulp of chil- expression of hepatic markers α-fetoprotein and albumin. The
dren’s primary, or deciduous, teeth. Shi, a pediatric dentist, was authors described the potentially considerable ramifications of
helping his daughter pull out a loose baby tooth when he this discovery: “To produce . . . tissue for transplantation, without
noticed residual pulp inside the tooth. The discovery that the feeder layers, and with the . . . recipient’s immunological pheno-
stem cells remained viable inside the tooth for hours after it type, is a . . . scientific hindrance. . . . The annual global 100 million
was de-rooted prompted him to consider harvestation. Shi human birth rate proposes umbilical cord blood as the largest

NEUROSURGERY VOLUME 64 | NUMBER 1 | JANUARY 2009 | 23


FARIN ET AL.

potential for clinical applica-


tions” (69, p 245).

2005: President Bush


Approves Federal Funds
for Restricted Stem Cell
Research
On December 20, 2005,
President George W. Bush for-
mally approv ed the use of
federal funds for research on
existing stem cell lines by
signing into law H.R. 2520,
the Stem Cell Therapeutic and
Research Act of 2005. The Act
created a new federal pro-
gram that promotes stem cell
research by re quiring cord
blood banks to donate to re-
searchers units not suitable
for transplantation because of
disease or size. The Act also
ex panded the current bone
marrow registry program to
also include cord blood (36).
Pres ident Bush’s decision
reflects widespread and high-
level recognition of the thera-
peutic potential of stem cells.
However, the decision to fund
only research on existing stem
cell lines and to severely
restrict federal funding for
embryonal research is viewed
by many as a significant neg-
ative position that reflects the
President’s personal religious
beliefs, and it led many scien-
tists to search for alternative
FIGURE 8. A–H, neural differentiation of stem cells from human exfoliated deciduous teeth (SHED). Immunocytochemical
pluripotent stem cell sources.
staining depicts SHED expressing the neural cell markers nestin, glial fibrillary acidic protein (GFAP), neurofilament-M
Partly in response to signifi-
(NFM), 2ⴕ,3ⴕ-cyclic nucleotide-3ⴕ-phosphodiesterase (CNPase), βIII-tubulin, glutamic acid decarboxylase (GAD), and neu-
ronal nuclei (NeuN). I, Western blot analysis confirmed that SHED expressed neural markers as described above. After 4 weeks cant global legislative and
of culture in the presence of B27 supplement, basic fibroblast growth factor, and epidermal growth factor, expression levels of ethical concerns complicating
βIII-tubulin, GAD, and NeuN were up-regulated when compared with regular culture conditions. However, expression lev- ES cell research, investigators
els of nestin, GFAP, CNPase, and NFM remained the same after the treatment. J–O, SHED may coexpress neuronal mark- more intensely sought out
ers including βIII-tubulin (green)/GAD (red) and βIII-tubulin (green)/NFM (red). The morphology of SHED showed elon- alternative means to derive
gated cell-cytoplasmic processes that sometimes coexpress neural markers (triangles) or only express individual neural marker pluripotent stem cells.
(arrows). P–S, toluidine blue (0.1%) staining depicting the altered morphology of SHED after induction with neural culture
medium (P and Q, arrows). Immunopositive staining for anti-MAP2 and anti-Tau antibodies on dendrites and axon (R and August 2006: Murine
S, arrows), respectively. Double-staining experiments showed that βIII-tubulin-positive cells were also detected in the same Pluripotent Stem Cells
field (R, triangle, green). T–W, SHED continued to express glial cell markers including nestin (red), CNPase (red), GFAP Generated from Fibroblasts
(red), and NFM (green) by immunocytostaining (from, Miura M, Gronthos S, Zhao M, Lu B, Fisher LW, Robey PG, Shi S: with Specific Factors
SHED: Stem cells from human exfoliated deciduous teeth. Proc Natl Acad Sci U S A 100:5807–5812, 2003 [74]).
Accordingly, in 2006, Taka-
hashi and Yamanaka (121)
untouched stem cell source. . . . CBEs are a viable . . . alternative demonstrated that differentiated adult fibroblasts can be “repro-
from embryonic stem cells . . . without ethical constraint and with grammed” backward or induced to become embryonic-like

24 | VOLUME 64 | NUMBER 1 | JANUARY 2009 www.neurosurgery-online.com


FOUNDATIONS AND LANDMARKS IN STEM CELL BIOLOGY

pluripotent stem cells. Transference of nuclear contents into June 2007: Mouse Fibroblasts Reprogrammed
oocytes resulted in cells engineered to express 4 defined factors, to Embryonic State
Oct3/4, Sox2, c-Myc, and Klf4, which are known to be impor- As a prelude to the same phenomenon soon thereafter
tant for maintaining the “stemness” of ES cells. Induction also observed in humans, research reported by 3 different groups
required ES cell culture conditions. These iPS cells revealed the showed that skin cells, or fibroblasts, can be reprogrammed to
morphology and growth properties of true ES cells and an embryonic state in mice (18). Shinya Yamanaka of Kyoto
expressed ES cell marker genes. Subcutaneous transplantation University, who pioneered the technique, had induced pluripo-
of the induced cells into nude mice resulted in teratomas. After tent cells from mouse fibroblasts in 2006 via retroviral transfer
injection into blastocysts, the induced cells contributed to mouse of 4 genes; resultant transcription factors facilitated the expres-
embryonic development. sion of additional genes that generated pluripotency. Although
November 2006: Mouse ES Cells Preserve the induced pluripotent cells had some characteristics of
Life in Mice with Liver Failure embryonic cells in that they could propagate continuously and
could form teratomas, they failed to produce a chimera mouse
Japanese scientists collaborating with the National Institutes when injected into a developing embryo. (A chimera would be
of Health successfully induced mouse ES cells to differentiate characterized by DNA from both the original embryo and the
into hepatocytes, forming an implantable bioartificial liver that induced pluripotent cells.) The 4 transcription factors used by
replaced aspects of physiologically normal liver function. The Yamanaka in 2006 reprogrammed less than 0.1% of the million
ES cell-derived hepatocytes expressed liver-specific genes, cells in a skin biopsy.
secreted albumin, and metabolized ammonia, lidocaine, and
diazepam. Treatment of 90% hepatectomized mice with an
A
implanted bioartificial liver seeded with ES cell-derived hepa-
tocytes improved liver function and prolonged survival,
whereas treatment with a bioartificial liver seeded with control
cells did not. The hope is to carry out the same results with
human stem cells (111).

January 2007: Discovery of Human Amniotic


Stem Cell Lines
Scientists at Wake Forest University, led by Dr. Anthony
Atala, and at Harvard University reported discovery of amni-
otic fluid-derived stem cells expressing both ES and adult stem
cell markers; embryonal destruction is precluded with this B
methodology (20). Undifferentiated cells doubled in 36 hours
and did not become tumorigenic, although they could give rise
to tissues of all 3 embryonic germ layers. After dozens of pop-
ulation doublings, long telomeres and normal karyotypes were
retained. Differentiated cells derived from amniotic fluid stem
cells included neuronal lineage cells, hepatic lineage cells, and
osteogenic lineage cells.
Dr. Robert Lanza, chief scientist at the stem cell company
Advanced Cell Technology, which later cloned the first human FIGURE 9. Clonal human amniotic fluid-derived stem (AFS) cells are
broadly multipotent. The clonality of AFS cells and differentiated cells was
embryo, stated that “ . . . this work represents a giant step for-
verified. A, reverse transcription-polymerase chain reaction (RT-PCR)
ward for stem cell research” (22a). Dr. George Daley, a Harvard
analysis of messenger ribonucleic acids for lineages indicated. U, control
University stem cell researcher, indicated that expectant par- undifferentiated cells; D, cells maintained under conditions to promote
ents may someday be able to store amniotic stem cells for osteogenic, myogenic, adipogenic, endothelial, hepatic, and neurogenic dif-
future tissue replacement in a sick child without fear of ferentiation. B, Southern blot analysis of inserted green fluorescent protein
immune rejection. However, he warned, “While they are fasci- (GFP) retroviral deoxyribonucleic acid (DNA) in differentiated cells from
nating subjects of study in their own right, they are not a sub- A; arrow indicates 4-kilobase pair (kbp) junction fragment in BamHI-
stitute for human ES cells, which allow scientists to address a digested DNA from undifferentiated cells of a GFP-positive subclone of AFS
host of other interesting questions in early human develop- cells (Lane 1) and the second round subclone (Lane 2) used for transcript
ment” (74a). Interestingly, some of the DNA retrieved from analysis after differentiation under adipogenic (Lane 3), endothelial (Lane
4), hepatic (Lane 5), osteogenic (Lane 6), myogenic (Lane 7), and neuro-
the amniotic stem cells contained Y chromosomes, indicating
genic (Lane 8) conditions (from, De Coppi P, Bartsch G Jr, Siddiqui MM,
that the cells came from male babies rather than the pregnant
Xu T, Santos CC, Perin L, Mostoslavsky G, Serre AC, Snyder EY, Yoo JJ,
mothers. Stem cells were reportedly easily extracted without Furth ME, Soker S, Atala A: Isolation of amniotic stem cell lines with
significant risk to mother or fetus; brain, liver, and bone tissue potential for therapy. Nat Biotechnol 25:100–106, 2007 [20]).
types were subsequently developed (Figs. 9–12).

NEUROSURGERY VOLUME 64 | NUMBER 1 | JANUARY 2009 | 25


FARIN ET AL.

A B A B

C C D

D
E F

FIGURE 11. Engraftment of neurogenically differentiated human AFS


cells in mouse brain. Differentiation was induced by incubation with NGF,
and cells were injected into brains of newborn mice. Samples shown were
obtained 1 month after injection. Red staining with antibody to a 65-kDa
FIGURE 10. Neurogenic differentiation of human AFS cells in culture. A, mitochondrial protein (perinuclear localization) reveals human cells. Blue
immunofluorescence staining for nestin (a neural marker) after 8 days in staining (4ⴕ,6-diamidino-2-phenylindole, DAPI) is a fluorescent stain that
the first stage of neurogenic differentiation. B, phase contrast micrograph binds strongly to DNA; superimposed image in D and insets of A, B, E, F
showing cells after the second stage of neurogenic differentiation under show cell nuclei. A, lateral ventricle; B, higher-magnification view of lat-
conditions biased for production of dopaminergic neurons; individual cells eral ventricle; C, periventricular area and hippocampus; D, same field as C,
with pyramidal morphology were assessed for potassium channels by volt- with DAPI staining superimposed; E, third ventricle; F, olfactory bulb
age clamping. C, RT-PCR showing expression of the housekeeping enzyme (from, De Coppi P, Bartsch G Jr, Siddiqui MM, Xu T, Santos CC, Perin L,
glyceraldehyde-3-phosphate dehydrogenase (Lane 1) and the neuronal G Mostoslavsky G, Serre AC, Snyder EY, Yoo JJ, Furth ME, Soker S, Atala
protein-coupled, inwardly rectifying K+ channel (GIRK2, Lane 2) after sec- A: Isolation of amniotic stem cell lines with potential for therapy. Nat
ond stage of neurogenic differentiation. M, size markers. Sizes of PCR Biotechnol 25:100–106, 2007 [20]).
products are shown. bp, base pairs. D, secretion of neurotransmitter glu-
tamic acid in response to potassium ions in cells after neurogenic differen-
tiation in the presence of nerve growth factor (NGF) (open bars) and after Konrad Hochedlinger of the Harvard Stem Cell Institute and
maintenance of undifferentiated cells in standard growth medium (closed Kathrin Plath (63) of the University of California, Los Angeles,
bars) (from, De Coppi P, Bartsch G Jr, Siddiqui MM, Xu T, Santos CC, used the same factors as Yamanaka did in 2007 and obtained
Perin L, Mostoslavsky G, Serre AC, Snyder EY, Yoo JJ, Furth ME, Soker similar results. These groups produced chimeric mice using
S, Atala A: Isolation of amniotic stem cell lines with potential for therapy. induced pluripotent cells that were isolated in this manner, and
Nat Biotechnol 25:100–106, 2007 [20]). the mice passed on induced pluripotent DNA to their offspring.
Jaenisch also produced embryos and fetuses whose cells were
Isolating cells that were successfully reprogrammed was derived entirely from induced pluripotent cells.
more efficiently accomplished in 2007 by inserting a gene for This June 2007 discovery was laudable for providing pluri-
antibiotic resistance that is activated only when proteins charac- potent stem cells that were genetically matched to individuals
teristic of stem cells are expressed. Antibiotics are used to elim- while minimizing ethical and technical dilemmas involved in
inate nonreprogrammed cells (83). A group led by Rudolf embryonal destruction or human cloning to otherwise obtain a
Jaenisch (136) at the Whitehead Institute for Biomedical genetic match. Whereas human cloning is limited by the num-
Research in Cambridge, MA, and a collaborative effort between ber of available eggs and technical difficulties, Yamanaka’s

26 | VOLUME 64 | NUMBER 1 | JANUARY 2009 www.neurosurgery-online.com


FOUNDATIONS AND LANDMARKS IN STEM CELL BIOLOGY

structed blastocysts expressed


A B C markers similar to those ex-
pressed by fertilized embryos.
The authors summarized, “Our
results represent a significant
breakthrough in elucidating
the role of nuclear remodeling
events in reprogramming fol-
lowing SCNT” (71, p 2232).
In 2004, cell nuclear transfer,
or cloning, was reportedly used
by Hwang Woo-Suk in Korea
D E F to produce a line of human ES
cells from fertilized human
oocytes. His work was sub-
sequently shown to be fabri-
cated.

October 2007: Nobel


Prize Awarded for
Engineering Knockout
Mice with ES Cells
The 2007 Nobel Prize for
Physiology or Medicine was
awarded to Mario Capecchi,
Martin Evans, and Oliver
FIGURE 12. Tissue engineered bone from human AFS cells. A, calcium deposition of human AFS cells maintained Smithies for their discoveries
in osteogenic differentiation medium in vitro was quantified by measuring calcium-cresolophthalein complex levels
of “principles for introducing
(solid line) and compared with that of undifferentiated (broken line) AFS cells. B, staining of control alginate/collagen
scaffold recovered 8 weeks after implantation in nu/nu mouse. C, staining of scaffold of AFS cells in alginate/collagen
specific gene modifications in
scaffold recovered 8 weeks after implantation; black staining indicates strong mineralization. D–F, micro-computed mice by the use of embryonic
tomographic scan of mouse 18 weeks after implantation of constructs. 䉴, region of implantation of control scaffold with- stem cells” and generating
out AFS cells; * and 䉫, scaffolds seeded with AFS cells. Close-up views of seeded scaffolds are indicated by symbols knockout mice (Fig. 13) (124).
(E, F) (from, De Coppi P, Bartsch G Jr, Siddiqui MM, Xu T, Santos CC, Perin L, Mostoslavsky G, Serre AC, Snyder The magnitude of their enor-
EY, Yoo JJ, Furth ME, Soker S, Atala A: Isolation of amniotic stem cell lines with potential for therapy. Nat mous contribution to medi-
Biotechnol 25:100–106, 2007 [20]). cine is challenging to summa-
rize adequately. Their efforts
method utilizes the most basic cells and simple laboratory tech- emphasize, in this most crucial context, the irreplaceable value
niques. Conceivably, if the same techniques were applied to of pluripotent ES cells.
humans, researchers could produce iPS cells from patients with Capecchi and Smithies initially noted that homologous recom-
Parkinson’s disease or diabetes for therapeutic cell replace- bination, or the natural exchange of DNA sequences within chro-
ment. However, the induced pluripotent cells cannot, as of mosome pairs to increase genetic variety, could be instigated
now, be used safely to generate genetically matched cells for between introduced DNA and endogenous chromosomes, result-
transplantation, as the technique carries a 20% risk of onco- ing in repair of defective genes. It appeared that any mammalian
genic potential in mice. gene could be targeted for specific modification by homologous
recombination. Evans then provided the crucial conduit to the
June 2007: First Successful Primate Stem Cell mouse germline so that DNA modifications would be inherited
Line Created via the ES cell. He observed that ES cells with normal karyotypes
Scientist Shoukhrat Mitalipov of Oregon Health & Science could be established from early mouse embryos. Embryos from
University reported the first successful creation of a primate one mouse strain were injected with ES cells from another mouse
stem cell line through somatic cell nuclear transfer (SCNT) (10, strain, resulting in mosaic embryos that were then carried to term
71). This technique requires cytoplast-mediated reprogramming by surrogate mothers. The mosaic offspring was subsequently
of the donor nucleus through nuclear remodeling. This triumph mated. ES cell-derived genes were then identified in pups, reflect-
had proven challenging during previous attempts at SCNT in ing Mendelian inheritance of genetic modifications. Evans began
primates; in this case, more than 20% of SCNT embryos recon- to modify the ES cells with retroviruses, which integrated their
structed with fetal fibroblasts progressed to blastocysts. Suc- genes into the embryonic chromosomes, generating the first gene-
cessful reprogramming was reflected in the fact that recon- targeted ES cells. Evans’ findings, along with Capecchi and

NEUROSURGERY VOLUME 64 | NUMBER 1 | JANUARY 2009 | 27


FARIN ET AL.

produce almost any variety of


DNA modification in the
mouse genome by gene target-
ing and thereby investigate
almost every feature of mam-
malian physiology. This potent
technical feat of gene targeting
in mice has already produced
more than 500 different mouse
models of human disease,
enabling investigators to con-
firm the roles of hundreds of
genes in mammalian embry-
onic and fetal development
including organogenesis and
body plan establishment, adult
physiology, aging, and disease
mechanisms. The models also
enable testing of the effects of
gene therapy. Today, gene tar-
geting has evolved so far as to
permit the introduction of tem-
porally regulated mutations as
well as organ-specific muta-
tions during development and
in the adult animal. As the
Nobel Committee aptly noted,
the birth of the knockout
mouse marked the beginning
of a new era in genetics.

November 2007: iPS Cells


Developed from
Adult Human Fibroblasts
Only a few months after
Yamanaka’s 2007 publication
(83), 2 similar articles by Taka-
hashi et al. (122) and Yu et al.
(143) showed that human iPS
cells could be generated from
mature human dermal fibro-
blasts. Through SCNT, trans-
acting factors of the mamma-
FIGURE 13. General strategy for gene targeting in mice (from, The Nobel Prize in Physiology or Medicine 2007. lian oocyte reprogram somatic
http://nobelprize.org/nobel_prizes/medicine/laureates/2007/press.html. Accessed May 1, 2008 [124]). nuclei to an undifferentiated
state. Human iPS cells were
Smithies’ demonstration that genes could be targeted by homolo- similar to human ES cells in morphology, karyotypes, prolifer-
gous recombination in cultured cells, led to the concept of homol- ation, surface antigens, and gene expression. In addition, iPS
ogous recombination in ES cells. Initial experiments were per- cells could differentiate into teratomas in vivo. This momen-
formed with the hprt gene for Lesch-Nyhan syndrome. Capecchi tous achievement demonstrated the feasibility of producing a
had developed a more advantageous tactic for targeting genes, pluripotent stem cell from almost any other human cell rather
termed positive-negative selection. The original documentation than necessitating an embryo or egg. As Takahashi et al. (122)
of homologous recombination in ES cells used to generate gene- pointed out, “Successful reprogramming of differentiated
targeted mice was published in 1989. human somatic cells into a pluripotent state would allow cre-
To date, more than 10 000 murine genes, or half the genes in the ation of patient- and disease-specific stem cells” (122, p 861)
mammalian genome, have been knocked out, with the capacity to with tremendous clinical applications in disease models, drug

28 | VOLUME 64 | NUMBER 1 | JANUARY 2009 www.neurosurgery-online.com


FOUNDATIONS AND LANDMARKS IN STEM CELL BIOLOGY

development, and transplantation medicine. However, the risk from the embryos. Richard Doerflinger, spokesman for the
of tumorigenesis resulting from retroviral gene transfer (Oct3/4, United States Conference of Catholic Bishops, criticized that the
Sox2, Klf4, and c-Myc in Yamanaka’s work and Oct4, Sox2, process still “involves creating human lives in the laboratory
NANOG, and Lin28 in Thomson’s work) requires more analysis. solely to destroy them for alleged benefit to others” (25).
Although there was no progress beyond the blastocyst stage, the
January 2008: Human ES Cell Lines Generated without process did enable all intended parents to successfully achieve
Embryo Destruction ongoing pregnancies with the oocytes retained for reproduc-
Advanced Cell Technology announced the development of 5 tive purposes.
human ES cell lines generated without embryonal destruction
(13). All previous human ES cell lines involved destruction of February 2008: Improved Generation of Mouse iPS Cells
embryos. These results have the potential to definitively end In February 2008, Yamanaka developed murine iPS cells that
the ethical debate surrounding the use of embryos to derive appeared to be more similar to ES cells than the previously
stem cells. developed induced pluripotent cells. Previously, iPS cells gener-
Single cells were removed from embryos using a technique ated from mouse and human fibroblasts required retroviral
similar to preimplantation genetic diagnosis. The biopsied transduction of 4 transcription factors. In their recent article,
embryos continued to develop normally to the blastocyst stage Aoi et al. (2) describe generation of iPS cells from adult mouse
and were then frozen. The isolated cells were cultured with the hepatocytes and gastric epithelial cells without the need for
use of a proprietary methodology recreating the optimal inner retroviral integration. Specifically, 4 transcription factors
cell mass developmental environment. This substantially (Oct3/4, Sox2, Klf4, and c-Myc) were introduced by retroviral
improved the efficiency of pluripotent stem cell derivation, vectors into hepatocytes or gastric epithelial cells. iPS cells were
achieving a rate comparable to the traditional approach of generated by germline transmission of induction genes, as
deriving stem cells from a blastocyst’s inner cell mass. The 5 judged from the presence of transgenes, without retroviral inte-
lines of stem cells maintained normal karyotypes and markers gration into specific sites. Few retroviral integration sites were
of pluripotency for up to 50 passages. They also differentiated noted at all, and no common retroviral integration sites were
into cell types of all 3 germ layers, including blood cells, neu- found among multiple clones. Instead, integration sites were
rons, myocytes, cartilage, and other cell types of therapeutic randomly distributed in multiple chromosomes. Each of the 4
significance. Human ES cell coculture was not essential to the factors were individually omitted to assess the effect on the gen-
derivation. The stem cell lines that were generated appear to eration of iPS cells. When Oct3/4, Sox2, or Klf4 were omitted,
have the same characteristics as other human ES cell lines, no iPS cell colonies emerged. c-Myc omission decreased colony
including expression of the same markers of pluripotency, self- numbers 20 to 40%, reflecting its minor role in the generation of
renewing capacity, and genetic stability. Robert Lanza, M.D., cells, compared with its role in generating iPS fibroblast cells.
Chief Scientific Officer at Advanced Cell Technology and senior Generation of iPS cells with gene transfer methods, or direct
author of the article, commented, “If the White House approves reprogramming of lineage-committed somatic cells, instead of
this new methodology, researchers could effectively double or by retroviral integration of induction genes into specific sites,
triple the number of stem cell lines available within a few diminishes tumorigenicity after transplantation into patients
months. Too many needless deaths continue to occur while this and is a major technical advance (2).
research is being held up. I hope the President will act now and An abridged timeline of the aforementioned significant
approve these stem cell lines quickly” (1a). events in stem cell science is illustrated in Figure 14.

January 2008: Development of Human Cloned Blastocysts


CONCLUSIONS
Scientists at Stemagen Corp. in La Jolla, CA, claimed develop-
ment of human cloned blastocysts after SCNT with adult fibrob- Although stem cell research has been applied to an extensive
lasts (25). First, oocytes obtained from egg donors who were 20 list of disorders (19, 42, 43, 50, 82, 84–86, 88, 96, 119), its true
to 24 years of age were enucleated. Then, SCNT embryos were long-term potential extends beyond any narrow discipline.
constructed using skin cells from 2 men. The cells were incu- Investigators are optimistic that, in the coming decades, stem
bated; cleavage and blastocyst development were observed. cells may be used successfully to battle medicine’s toughest
Unfortunately, no stem cells were extracted, but DNA was con- foes: malignant cancer, cardiovascular disease, and central nerv-
firmed to match the male donors. Blastocysts were also obtained ous system injuries and deficits. Those in need of transplants
from parthenogenetically activated oocytes. In 2005, British sci- hope newly engineered organs will one day obviate the need to
entists reported using human ES cells to produce a cloned wait for organs. Perhaps most interestingly, the embryogenetic
embryo, but no stem cells were extracted then, either. This study mishaps leading to birth defects will be better characterized by
demonstrates, for the first time, that SCNT can be used to gen- studying stem cells. Immediate future efforts must include as
erate cloned human blastocysts using differentiated adult donor their focus safeguarding humans against any tumorigenic
nuclei that are reprogrammed by human oocytes. Dr. Samuel potential from stem cell transplantation.
Wood, coauthor of the article and Chief Executive of Stemagen, Although stem cell research is still in its infancy, its imagina-
reports that he is now attempting to produce stem cell lines ble clinical applications seem almost limitless. These potentially

NEUROSURGERY VOLUME 64 | NUMBER 1 | JANUARY 2009 | 29


FARIN ET AL.

FIGURE 14. Timeline of


significant events in stem
cell science. SCID, severe
combined immunodefi-
ciency; NIH, National
Institutes of Health.

30 | VOLUME 64 | NUMBER 1 | JANUARY 2009 www.neurosurgery-online.com


FOUNDATIONS AND LANDMARKS IN STEM CELL BIOLOGY

immortal cells are believed to be the key to understanding the 16. Connor DA: Mouse embryonic stem (ES) cell culture. Curr Protoc Mol Biol
mysterious processes of human embryogenesis, development, 23:23.3, 2001.
17. Cowan CA, Klimanskaya I, McMahon J, Atienza J, Witmyer J, Zucker JP,
aging, and disease. The ability to generate any type of cell in the
Wang S, Morton CC, McMahon AP, Powers D, Melton DA: Derivation of
body theoretically allows stem cells to repair any damaged or embryonic stem-cell lines from human blastocysts. N Engl J Med 350:1353–
inadequate tissue and conceivably to cure disease. Patients and 1356, 2004.
physicians alike eagerly anticipate the safe, effective, and ethi- 18. Cyranoski D: Simple switch turns cells embryonic. Nature 447:618–619, 2007.
cal clinical use of stem cells for therapeutic applications. 19. De Bari C, Dell’Accio F, Vandenabeele F, Vermeesch JR, Raymackers JM,
Luyten FP: Skeletal muscle repair by adult human mesenchymal stem cells
from synovial membrane. J Cell Biol 160:909–918, 2003.
Disclosure 20. De Coppi P, Bartsch G Jr, Siddiqui MM, Xu T, Santos CC, Perin L, Mosto-
The authors have no personal financial or institutional interest in any of the slavsky G, Serre AC, Snyder EY, Yoo JJ, Furth ME, Soker S, Atala A: Isolation
drugs, materials, or devices described in this article. of amniotic stem cell lines with potential for therapy. Nat Biotechnol
25:100–106, 2007.
21. Dooren LJ, de Vries MJ, van Bekkum DW, Cleton FJ, de Koning J: Sex-linked
REFERENCES thymic epithelial hypoplasia in two siblings. Attempt at treatment by trans-
plantation with fetal thymus and adult bone marrow. J Pediatr 72:51–62,
1. Amit M, Itskovitz-Eldor J: Feeder-free culture of human embryonic stem 1968.
cells. Methods Enzymol 420:37–49, 2006. 22. Draper JS, Smith K, Gokhale P, Moore HD, Maltby E, Johnson J, Meisner L,
1a. Anonymous: Advanced cell technology announces creation of human Zwaka TP, Thomson JA, Andrews PW: Recurrent gain of chromosomes 17q
embryonic stem cell lines without the destruction of embryos. http://advanced and 12 in cultured human embryonic stem cells. Nat Biotechnol 22:53–54,
cell.com/press-release/advanced-cell-technology-announces-creation-of- 2004.
human-embryonic-stem-cell-lines-without-the-destruction-of-embryos. 22a. Elias P: ‘Giant step’ for stem cell research: Amniotic fluid a promising source.
Accessed 10/27/2008. http://aproundtable.org/news.cfm?NEWS_ID=16158.issuecode. Accessed
2. Aoi T, Yae K, Nakagawa M, Ichisaka T, Okita K, Takahashi K, Chiba T, 10/27/2008.
Yamanaka S: Generation of pluripotent stem cells from adult mouse liver and 23. Evans MJ, Kaufman MH: Establishment in culture of pluripotential cells
stomach cells. Science 321:699–702, 2008. from mouse embryos. Nature 292:154–156, 1981.
3. Armstrong L, Lako M, Lincoln J, Cairns PM, Hole N: MTert expression cor- 24. Expert Reviews in Molecular Medicine. http://expertreviews.org. Accessed
relates with telomerase activity during the differentiation of murine embry- 5/1/2008.
onic stem cells. Mech Dev 97:109–116, 2000. 25. French AJ, Adams CA, Anderson LS, Kitchen JR, Hughes MR, Wood SH:
4. Avilion AA, Nicolis SK, Pevny LH, Perez L, Vivian N, Lovell-Badge R: Development of human cloned blastocysts following somatic cell nuclear
Multipotent cell lineages in early mouse development depend on SOX2 func- transfer with adult fibroblasts. Stem Cells 26:485–493, 2008.
tion. Genes Dev 17:126–140, 2003. 26. Friedenstein AJ, Deriglasova UF, Kulagina NN, Panasuk AF, Rudakowa SF,
5. Barrilleaux B, Phinney DG, Prockop DJ, O’Connor KC: Review: Ex vivo engi- Luriá EA, Ruadkow IA: Precursors for fibroblasts in different populations of
neering of living tissues with adult stem cells. Tissue Eng 12:3007–3019, hematopoietic cells as detected by the in vitro colony assay method. Exp
2006. Hematol 2:83–92, 1974.
6. Beckmann J, Scheitza S, Wernet P, Fischer JC, Giebel B: Asymmetric cell divi- 27. Friedenstein AJ, Gorskaja JF, Kulagina NN: Fibroblast precursors in normal
sion within the human hematopoietic stem and progenitor cell compart- and irradiated mouse hematopoietic organs. Exp Hematol 4:267–274, 1976.
ment: Identification of asymmetrically segregating proteins. Blood 109:5494– 28. Gardner RL: Stem cells: Potency, plasticity and public perception. J Anat
5501, 2007. 200:277–282, 2002.
7. Bongso A, Lee EH: Stem cells: Their definition, classification and sources, in 29. Gatti RA, Meuwissen HJ, Allen HD, Hong R, Good RA: Immunological
Bongso A, Lee EH (eds): Stem Cells: From Bench to Bedside. Singapore, World reconstitution of sex-linked lymphopenic immunological deficiency. Lancet
Scientific, 2005, pp 1–13. 2:1366–1369, 1968.
8. Boyer LA, Lee TI, Cole MF, Johnstone SE, Levine SS, Zucker JP, Guenther 30. Genbacev O, Krtolica A, Zdravkovic T, Brunette E, Powell S, Nath A, Caceres
MD, Kumar RM, Murray HL, Jenner RG, Gifford DK, Jaenisch R, Young RA: E, McMaster M, McDonagh S, Li Y, Mandalam R, Lebkowski J, Fisher SJ:
Core transcriptional regulatory circuitry in human embryonic stem cells. Serum-free derivation of human embryonic stem cell lines on human placen-
Cell 122:947–956, 2005. tal fibroblast feeders. Fertil Steril 83:1517–1529, 2005.
9. Bryja V, Bonilla S, Cajánek L, Parish CL, Schwartz CM, Luo Y, Rao MS, 31. Gertow K, Cedervall J, Unger C, Szöke K, Blennow E, Imreh MP, Ahrlund-
Arenas E: An efficient method for the derivation of mouse embryonic stem Richter L: Trisomy 12 in HESC leads to no selective in vivo growth advantage
cells. Stem Cells 24:844–849, 2006. in teratomas, but induces an increased abundance of renal development.
10. Byrne JA, Pedersen DA, Clepper LL, Nelson M, Sanger WG, Gokhale S, Wolf J Cell Biochem 100:1518–1525, 2007.
DP, Mitalipov SM: Producing primate embryonic stem cells by somatic cell 32. Gimble JM, Katz AJ, Bunnell BA: Adipose-derived stem cells for regenerative
nuclear transfer. Nature 450:497–502, 2007. medicine. Circ Res 100:1249–1260, 2007.
11. Chambers I: http://www.sciencedaily.com/releases/2003/06/030602024530. 33. Ginis I, Luo Y, Miura T, Thies S, Brandenberger R, Gerecht-Nir S, Amit M,
htm. Accessed 10/27/2008. Hoke A, Carpenter MK, Itskovitz-Eldor J, Rao MS: Differences between
11a. Chambers I, Colby D, Robertson M, Nichols J, Lee S, Tweedie S, Smith A: human and mouse embryonic stem cells. Dev Biol 269:360–380, 2004.
Functional expression cloning of Nanog, a pluripotency sustaining factor in 34. Green RM: The Human Embryo Research Debates: Bioethics in the Vortex of
embryonic stem cells. Cell 113:643–655, 2003. Controversy. New York, Oxford University Press, 2001, ed 1.
12. Christensen B: World’s first artificial human liver grown in lab. http:// 35. Gropp M, Reubinoff B: Lentiviral vector-mediated gene delivery into human
livescience.com/health/061031_artif_liver.html. Accessed 5/1/2008. embryonic stem cells. Methods Enzymol 420:64–81, 2006.
13. Chung Y, Klimanskaya I, Becker S, Li T, Maserati M, Lu SJ, Zdravkovic T, Ilic 36. H.R. 2520 [109th]: Stem Cell Therapeutic and Research Act of 2005.
D, Genbacev O, Fisher S, Krtolica A, Lanza R: Human embryonic stem cell http://govtrack.us/congress/bill.xpd?bill5h109-2520. Accessed May 1,
lines generated without embryo destruction. Cell Stem Cell 2:113–117, 2008. 2008.
14. Cibelli JB, Kiessling AA, Cunniff K, Richards C, Lanza RP, West MD: Somatic 37. Hanna LA, Foreman RK, Tarasenko IA, Kessler DS, Labosky PA: Require-
cell nuclear transfer in humans: Pronuclear and early embryonic develop- ment for Foxd3 in maintaining pluripotent cells of the early mouse embryo.
ment. E-Biomed: J Regenerative Medicine 2:25–31, 2001. Genes Dev 16:2650–2661, 2002.
15. Cibelli JB, Lanza RP, West MD, Ezzell C: The first human cloned embryo. Sci 38. Henderson JK, Draper JS, Baillie HS, Fishel S, Thomson JA, Moore H,
Am 286:44–51, 2002. Andrews PW: Preimplantation human embryos and embryonic stem cells

NEUROSURGERY VOLUME 64 | NUMBER 1 | JANUARY 2009 | 31


FARIN ET AL.

show comparable expression of stage-specific embryonic antigens. Stem 56. Liu X, Wu H, Loring J, Hormuzdi S, Disteche CM, Bornstein P, Jaenisch R:
Cells 20:329–337, 2002. Trisomy eight in ES cells is a common potential problem in gene targeting
39. Hoffman LM, Carpenter MK: Characterization and culture of human embry- and interferes with germ line transmission. Dev Dyn 209:85–91, 1997.
onic stem cells. Nature Biotech 23:699–708, 2005. 57. Liver cells grown from cord blood. http://news.bbc.co.uk/2/hi/health/
40. International Stem Cell Initiative, Adewumi O, Aflatoonian B, Ahrlund- 6101420.stm. Accessed 5/1/2008.
Richter L, Amit M, Andrews PW, Beighton G, Bello PA, Benvenisty N, 58. Loebel DA, Watson CM, De Young RA, Tam PP: Lineage choice and differ-
Berry LS, Bevan S, Blum B, Brooking J, Chen KG, Choo AB, Churchill GA, entiation in mouse embryos and embryonic stem cells. Dev Biol 264:1–14,
Corbel M, Damjanov I, Draper JS, Dvorak P, Emanuelsson K, Fleck RA, 2003.
Ford A, Gertow K, Gertsenstein M, Gokhale PJ, Hamilton RS, Hampl A, 59. Longo L, Bygrave A, Grosveld FG, Pandolfi PP: The chromosome make-up
Healy LE, Hovatta O, Hyllner J, Imreh MP, Itskovitz-Eldor J, Jackson J, of mouse embryonic stem cells is predictive of somatic and germ cell chi-
Johnson JL, Jones M, Kee K, King BL, Knowles BB, Lako M, Lebrin F, maerism. Transgenic Res 6:321–328, 1997.
Mallon BS, Manning D, Mayshar Y, McKay RD, Michalska AE, Mikkola M, 60. Lu J, Hou R, Booth CJ, Yang SH, Snyder M: Defined culture conditions of
Mileikovsky M, Minger SL, Moore HD, Mummery CL, Nagy A, Nakatsuji N, human embryonic stem cells. Proc Natl Acad Sci U S A 103:5688–5693, 2006.
O’Brien CM, Oh SK, Olsson C, Otonkoski T, Park KY, Passier R, Patel H, 61. Ludwig TE, Bergendahl V, Levenstein ME, Yu J, Probasco MD, Thomson JA:
Patel M, Pedersen R, Pera MF, Piekarczyk MS, Pera RA, Reubinoff BE, Feeder-independent culture of human embryonic stem cells. Nat Methods
Robins AJ, Rossant J, Rugg- Gunn P, Schulz TC, Semb H, Sherrer ES, 3:637–646, 2006.
Siemen H, Stacey GN, Stojkovic M, Suemori H, Szatkiewicz J, Turetsky T, 62. Ludwig TE, Levenstein ME, Jones JM, Berggren WT, Mitchen ER, Frane JL,
Tuuri T, van den Brink S, Vintersten K, Vuoristo S, Ward D, Weaver TA, Crandall LJ, Daigh CA, Conard KR, Piekarczyk MS, Llanas RA, Thomson JA:
Young LA, Zhang W: Characterization of human embryonic stem cell lines Derivation of human embryonic stem cells in defined conditions. Nat
by the International Stem Cell Initiative. Nat Biotechnol 25:803–816, 2007. Biotechnol 24:185–187, 2006.
41. Jiang Y, Jahagirdar BN, Reinhardt RL, Schwartz RE, Keene CD, Ortiz- 63. Maherali N, Sridharan R, Xie W, Utikal J, Eminli S, Arnold K, Stadtfeld M,
Gonzalez XR, Reyes M, Lenvik T, Lund T, Blackstad M, Du J, Aldrich Yachechko R, Tchieu J, Jaenisch R, Plath K, Hochedlinger K: Directly repro-
S, Lisberg A, Low WC, Largaespada DA, Verfaillie CM: Pluripotency of mes- grammed fibroblasts show global epigenetic remodeling and widespread
enchymal stem cells derived from adult marrow. Nature 418:41–49, 2002. tissue contribution. Cell Stem Cell 7:55–70, 2007.
42. Kajstura J, Leri A, Finato N, Di Loreto C, Beltrami CA, Anversa P: Myocyte 64. Martin GR: Isolation of a pluripotent cell line from early mouse embryos cul-
proliferation in end-stage cardiac failure in humans. Proc Natl Acad Sci tured in medium conditioned by teratocarcinoma stem cells. Proc Natl Acad
U S A 95:8801–8805, 1998. Sci U S A 78:7634–7638, 1981.
43. Kale S, Karihaloo A, Clark PR, Kashgarian M, Krause DS, Cantley LG: Bone 65. McCulloch EA: Stem cell renewal and determination during clonal expansion
in normal and leukaemic haemopoiesis. Cell Prolif 26:399–425, 1993.
marrow stem cells contribute to repair of the ischemically injured renal
66. McCulloch EA, Till JE: Effects of short-term culture on populations of hemo-
tubule. J Clin Invest 112:42–49, 2003.
poietic progenitor cells from mouse marrow. Cell Tissue Kinet 4:11–20, 1971.
44. Kaufman MH, Robertson EJ, Handyside AH, Evans MJ: Establishment of
67. McCulloch EA, Till JE: Regulatory mechanisms acting on hemopoietic stem
pluripotential cell lines from haploid mouse embryos. J Embryol Exp
cells: Some clinical implications. Am J Pathol 65:601–619, 1971.
Morphol 73:249–261, 1983.
68. McCulloch EA, Till JE: Perspectives on the properties of stem cells. Nat Med
45. Klimanskaya I, McMahon J: Approaches for derivation and maintenance of
11:1026–1028, 2005.
human ES cells: Detailed procedures and alternatives, in Lanza R, Weissman
69. McGuckin CP, Forraz N, Baradez MO, Navran S, Zhao J, Urban R, Tilton R,
I, Thomson J, Pederson R, Hogan B, Gearhart J, Blau H, Melton D, Moore M,
Denner L: Production of stem cells with embryonic characteristics from
Verfaillie C, Thomas ED, West M (eds): Handbook of Stem Cells. Amsterdam,
human umbilical cord blood. Cell Prolif 38:245–255, 2005.
Academic, 2004, vol 1, pp 437–449.
70. Menendez P, Wang L, Bhatia M: Genetic manipulation of human embryonic
46. Klimanskaya I, Chung Y, Meisner L, Johnson J, West MD, Lanza R: Human
stem cells: A system to study early human development and potential ther-
embryonic stem cells derived without feeder cells. Lancet 365:1636–1641,
apeutic applications. Curr Gene Ther 5:375–385, 2005.
2005.
71. Mitalipov SM, Zhou Q, Byrne JA, Ji WZ, Norgren RB, Wolf DP: Reprogram-
47. Klimanskaya I, Rosenthal N, Lanza R: Derive and conquer: Sourcing and
ming following somatic cell nuclear transfer in primates is dependent upon
differentiating stem cells for therapeutic applications. Nat Rev Drug Discov
nuclear remodeling. Hum Reprod 22:2232–2242, 2007.
7:131–142, 2008.
72. Mitalipova MM, Rao RR, Hoyer DM, Johnson JA, Meisner LF, Jones KL,
48. Kondoh G, Yamamoto Y, Yoshida K, Suzuki Y, Osuka S, Nakano Y, Morita T, Dalton S, Stice SL: Preserving the genetic integrity of human embryonic stem
Takeda J: Easy assessment of ES cell clone potency for chimeric development cells. Nat Biotechnol 23:19–20, 2005.
and germ-line competency by an optimized aggregation method. J Biochem 73. Mitsui K, Tokuzawa Y, Itoh H, Segawa K, Murakami M, Takahashi K,
Biophys Methods 39:137–142, 1999. Maruyama M, Maeda M, Yamanaka S: The homeoprotein Nanog is required for
49. Krause DS, Theise ND, Collector MI, Henegariu O, Hwang S, Gardner R, maintenance of pluripotency in mouse epiblast and ES cells. Cell 113:631–642,
Neutzel S, Sharkis SJ: Multi-organ, multi-lineage engraftment by a single 2003.
bone marrow-derived stem cell. Cell 105:369–377, 2001. 74. Miura M, Gronthos S, Zhao M, Lu B, Fisher LW, Robey PG, Shi S: SHED:
50. LaBarge MA, Blau HM: Biological progression from adult bone marrow to Stem cells from human exfoliated deciduous teeth. Proc Natl Acad Sci
mononucleate muscle stem cell to multinucleate muscle fiber in response to U S A 100:5807–5812, 2003.
injury. Cell 111:589–601, 2002. 74a. Moreno JD, Berger S: Newly discovered stem cells will not replace embry-
51. Lagasse E, Connors H, Al-Dhalimy M, Reitsma M, Dohse M, Osborne L, onic stem cells. http://www.americanprogressaction.org/issues/2007/
Wang X, Finegold M, Weissman IL, Grompe M: Purified hematopoietic stem amniotic_stem_cells.html. Accessed 10/27/2008.
cells can differentiate into hepatocytes in vivo. Nat Med 6:1229–1234, 2000. 75. Nagy A, Vintersten K: Murine embryonic stem cells. Methods Enzymol
52. Lakshmipathy U, Pelacho B, Sudo K, Linehan JL, Coucouvanis E, Kaufman 418:3–21, 2006.
DS, Verfaillie CM: Efficient transfection of embryonic and adult stem cells. 76. National Center for Biotechnology Information. http://www.ncbi.nlm.nih.
Stem Cells 22:531–543, 2004. gov. Accessed 5/1/2008.
53. Lanza RP, Cibelli JB, West MD: Human therapeutic cloning. Nat Med 77. Nichols J, Chambers I, Smith A: Derivation of germline competent embryonic
5:975–977, 1999. stem cells with a combination of interleukin-6 and soluble interleukin-6
54. Lanza RP, Cibelli JB, West MD: Prospects for the use of nuclear transfer in receptor. Exp Cell Res 215:237–239, 1994.
human transplantation. Nat Biotechnol 17:1171–1174, 1999. 78. Nichols J, Zevnik B, Anastassiadis K, Niwa H, Klewe-Nebenius D, Chambers
55. Lei T, Jacob S, Ajil-Zaraa I, Dubuisson JB, Irion O, Jaconi M, Feki A: Xeno-free I, Schöler H, Smith A: Formation of pluripotent stem cells in the mammalian
derivation and culture of human embryonic stem cells: Current status, prob- embryo depends on the POU transcription factor Oct4. Cell 95:379–391, 1998.
lems and challenges. Cell Res 17:682–688, 2007. 79. NIH Stem Cell Information. http://stemcells.nih.gov. Accessed 5/1/2008.

32 | VOLUME 64 | NUMBER 1 | JANUARY 2009 www.neurosurgery-online.com


FOUNDATIONS AND LANDMARKS IN STEM CELL BIOLOGY

80. Niho Y, Till JE, McCulloch EA: Granulopoietic progenitors in suspension 102. Robertson EJ, Evans MJ, Kaufman MH: X-chromosome instability in pluripo-
culture: A comparison of stimulatory cells and conditioned media. Blood tential stem cell lines derived from parthenogenetic embryos. J Embryol Exp
45:811–821, 1975. Morphol 74:297–309, 1983.
81. Niwa H, Miyazaki J, Smith AG: Quantitative expression of Oct-3/4 defines 103. Rogers MB, Hosler BA, Gudas LJ: Specific expression of a retinoic acid-
differentiation, dedifferentiation or self-renewal of ES cells. Nat Genet regulated, zinc-finger gene, Rex-1, in preimplantation embryos, trophoblast
24:372–376, 2000. and spermatocytes. Development 113:815–824, 1991.
82. Norol F, Merlet P, Isnard R, Sebillon P, Bonnet N, Cailliot C, Carrion C, 104. Rossant J: Stem cells and lineage development in the mammalian blastocyst.
Ribeiro M, Charlotte F, Pradeau P Mayol JD, Peinnequin A, Drouet M, Safsafi Reprod Fertil Dev 19:111–118, 2007.
K, Vernant JP, Herodin F: Influence of mobilized stem cells on myocardial 105. Schnieke AE, Kind AJ, Ritchie WA, Mycock K, Scott AR, Ritchie M, Wilmut I,
infarct repair in a nonhuman primate model. Blood 102:4361–4368, 2003. Colman A, Campbell KH: Human factor IX transgenic sheep produced by
83. Okita K, Ichisaka T, Yamanaka S: Generation of germline-competent induced transfer of nuclei from transfected fetal fibroblasts. Science 278:2130–2133, 1997.
pluripotent stem cells. Nature 448:313–317, 2007. 106. Schuldiner M, Yanuka O, Itskovitz-Eldor J, Melton DA, Benvenisty N: Effects
84. Orlic D, Kajstura J, Chimenti S, Jakoniuk I, Anderson SM, Li B, Pickel J, of eight growth factors on the differentiation of cells derived from human
McKay R, Nadal-Ginard B, Bodine DM, Leri A, Anversa P: Bone marrow embryonic stem cells. Proc Natl Acad Sci U S A 97:11307–11312, 2000.
cells regenerate infarcted myocardium. Nature 410:701–705, 2001. 107. Shi S, Gronthos S: Perivascular niche of postnatal mesenchymal stem cells in
85. Orlic D, Kajstura J, Chimenti S, Limana F, Jakoniuk I, Quaini F, Nadal-Ginard human bone marrow and dental pulp. J Bone Miner Res 18:696–704, 2003.
B, Bodine DM, Leri A, Anversa P: Mobilized bone marrow cells repair the 108. Simón C, Escobedo C, Valbuena D, Genbacev O, Galan A, Krtolica A, Asensi
infarcted heart, improving function and survival. Proc Natl Acad Sci U S A A, Sánchez E, Esplugues J, Fisher S, Pellicer A: First derivation in Spain of
98:10344–10349, 2001. human embryonic stem cell lines: Use of long-term cryopreserved embryos
86. Osawa M, Hanada K, Hamada H, Nakauchi H: Long-term lymphohe- and animal-free conditions. Fertil Steril 83:246–249, 2005.
matopoietic reconstitution by a single CD34-low/negative hematopoietic 109. Smith AG: Embryo-derived stem cells: Of mice and men. Annu Rev Cell
stem cell. Science 273:242–245, 1996. Dev Biol 17:435–462, 2001.
87. Palmieri SL, Peter W, Hess H, Schöler HR: Oct-4 transcription factor is differen- 110. Song X, Zhu C, Doan C, Xie T: Germline stem cells anchored by adherens
tially expressed in the mouse embryo during establishment of the first two junctions in the Drosophila ovary niches. Science 296:1855–1857, 2002.
extraembryonic cell lineages involved in implantation. Dev Biol 166:259–267, 111. Soto-Gutiérrez A, Kobayashi N, Rivas-Carrillo JD, Navarro-Alvarez N, Zhao
1994. D, Okitsu T, Noguchi H, Basma H, Tabata Y, Chen Y, Tanaka K, Narushima
88. Parker MH, Seale P, Rudnicki MA: Looking back to the embryo: Defining M, Miki A, Ueda T, Jun HS, Yoon JW, Lebkowski J, Tanaka N, Fox IJ: Reversal
transcriptional networks in adult myogenesis. Nat Rev Genet 4:497–507, of mouse hepatic failure using an implanted liver-assist device containing ES
2003. cell-derived hepatocytes. Nat Biotechnol 24:1412–9, 2006.
89. Pera MF, Reubinoff B, Trounson A: Human embryonic stem cells. J Cell Sci 112. Spradling A, Drummond-Barbosa D, Kai T: Stem cells find their niche.
113:5–10, 2000. Nature 414:98–104, 2001.
90. Pereira RF, Halford KW, O’Hara MD, Leeper DB, Sokolov BP, Pollard MD, 113. Staal FJ, Clevers HC: WNT signalling and haematopoiesis: A WNT-WNT sit-
Bagasra O, Prockop DJ: Cultured adherent cells from marrow can serve as uation. Nat Rev Immunol 5:21–30, 2005.
long-lasting precursor cells for bone, cartilage, and lung in irradiated mice. 114. Stamm C, Westphal B, Kleine HD, Petzsch M, Kittner C, Klinge H,
Proc Natl Acad Sci U S A 92:4857–4861, 1995. Schümichen C, Nienaber CA, Freund M, Steinhoff G: Autologous bone-
91. Petersen BE, Bowen WC, Patrene KD, Mars WM, Sullivan AK, Murase N, marrow stem-cell transplantation for myocardial regeneration. Lancet
Boggs SS, Greenberger JS, Goff JP: Bone marrow as a potential source of 361:45–46, 2003.
hepatic oval cells. Science 284:1168–1170, 1999. 115. Deleted in proof.
92. Polesskaya A, Seale P, Rudnicki MA: Wnt signaling induces the myogenic 116. Stemple DL, Anderson DJ: Isolation of a stem cell for neurons and glia from
specification of resident CD45+ adult stem cells during muscle regeneration. the mammalian neural crest. Cell 71:973–985, 1992.
Cell 113:841–852, 2003. 117. Stojkovic P, Lako M, Stewart R, Przyborski S, Armstrong L, Evans J, Murdoch
93. Price GB, Senn JS, McCulloch EA, Till JE: Heterogeneity of molecules with A, Strachan T, Stojkovic M: An autogeneic feeder cell system that efficiently
low molecular weight isolated from media conditioned by human leukocytes supports growth of undifferentiated human embryonic stem cells. Stem
and capable of stimulating colony formation by human granulopoietic pro- Cells 23:306–314, 2005.
genitor cells. J Cell Physiol 84:383–396, 1974. 118. Stojkovic M, Lako M, Stojkovic P, Stewart R, Przyborski S, Armstrong L,
94. Priller J, Persons DA, Klett FF, Kempermann G, Kreutzberg GW, Dirnagl U: Evans J, Herbert M, Hyslop L, Ahmad S, Murdoch A, Strachan T: Derivation
Neogenesis of cerebellar Purkinje neurons from gene-marked bone marrow of human embryonic stem cells from day-8 blastocysts recovered after three-
cells in vivo. J Cell Biol 155:733–738, 2001. step in vitro culture. Stem Cells 22:790–797, 2004.
95. Prindull G, Prindull B, Meulen N: Haematopoietic stem cells (CFUc) in 119. Strauer BE, Brehm M, Zeus T, Köstering M, Hernandez A, Sorg RV, Kögler G,
human cord blood. Acta Paediatr Scand 67:413–416, 1978. Wernet P: Repair of infarcted myocardium by autologous intracoronary
96. Prockop DJ, Gregory CA, Spees JL: One strategy for cell and gene therapy: mononuclear bone marrow cell transplantation in humans. Circulation
Harnessing the power of adult stem cells to repair tissues. Proc Natl Acad Sci 106:1913–1918, 2002.
U S A 100 [Suppl 1]:11917–11923, 2003. 120. Suzuki H, Kamada N, Ueda O, Jishage K, Kurihara Y, Kurihara H, Terauchi
97. Pyle AD, Lock LF, Donovan PJ: Neurotrophins mediate human embryonic Y, Azuma S, Kadowaki T, Kodama T, Yazaki Y, Toyoda Y: Germ-line contri-
stem cell survival. Nat Biotechnol 24:344–350, 2006. bution of embryonic stem cells in chimeric mice: Influence of karyotype and
98. Rao M: Conserved and divergent paths that regulate self-renewal in mouse in vitro differentiation ability. Exp Anim 46:17–23, 1997.
and human embryonic stem cells. Dev Biol 275:269–286, 2004. 121. Takahashi K, Yamanaka S: Induction of pluripotent stem cells from mouse
99. Ratajczak MZ, Machalinski B, Wojakowski W, Ratajczak J, Kucia M: A embryonic and adult fibroblast cultures by defined factors. Cell 126:663–676,
hypothesis for an embryonic origin of pluripotent Oct-4(+) stem cells in adult 2006.
bone marrow and other tissues. Leukemia 21:860–867, 2007. 122. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K,
100. Reubinoff BE, Pera MF, Fong CY, Trounson A, Bongso A: Embryonic stem cell Yamanaka S: Induction of pluripotent stem cells from adult human fibrob-
lines from human blastocysts: Somatic differentiation in vitro. Nat Biotechnol lasts by defined factors. Cell 131:861–872, 2007.
18:399–404, 2000. 123. The Multi-Dimensional Human Embryo. http://embryo.soad.umich.edu.
101. Reyer RW, Woolfitt RA, Withersty LT: Stimulation of lens regeneration from Accessed May 1, 2008.
the newt dorsal iris when implanted into the blastema of the regenerating 124. The Nobel Prize in Physiology or Medicine 2007. http://nobelprize.org/
limb. Dev Biol 32:258–281, 1973. nobel_prizes/medicine/laureates/2007/press.html. Accessed May 1, 2008.

NEUROSURGERY VOLUME 64 | NUMBER 1 | JANUARY 2009 | 33


FARIN ET AL.

125. Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, point out that the thorough evaluation of this topic will include the his-
Marshall VS, Jones JM: Embryonic stem cell lines derived from human blas- tory, scientific proofs of CNS neurogenesis, as well as the application of
tocysts. Science 282:1145–1147, 1998. this technology not only to stem cells but also to the derivation and
126. Till JE: Cellular diversity in the blood-forming system. Am Sci 69:522–527, 1981. control of function of other adult cells; finally, the article presents a dis-
127. Till JE: Stem cells in differentiation and neoplasia. J Cell Physiol Suppl
cussion of the philosophical and ethical issues that surround this topic.
1:3–11, 1982.
128. Till JE, McCulloch EA: Hemopoietic stem cell differentiation. Biochim The work is still in its very early stages, but this review suggests that
Biophys Acta 605:431–459, 1980. exciting work in associated areas, such as cloning and knockout mice,
129. Till JE, McCulloch EA, Siminovitch L: A stochastic model of stem cell prolif- has already completely changed the way we assess the pathophysiol-
eration, based on the growth of spleen colony-forming cells. Proc Natl Acad ogy of disorders. Also, it highlights the use of stem cell techniques to
Sci U S A 51:29–36, 1964. delineate causes of neoplasia in the CNS. We look forward to partici-
130. Till JE, Price GB, Lan S, McCulloch EA: Factors affecting the differentiation pating as a discipline in the development of this potential evolution in
of blood cells. Symp Soc Dev Biol 35:133–147, 1978. diagnosis, pathophysiology, and therapeutic advances.
131. Till JE, Price GB, Mak TW, McCulloch EA: Regulation of blood cell differen-
tiation. Fed Proc 34:2279–2284, 1975. Robert J. Dempsey
132. United States Conference of Catholic Bishops, Pro-Life Activities, Practical Madison, Wisconsin
Problems with Embryonic Stem Cells. http://www.usccb.org/prolife/
issues/bioethic/stemcell/obstacles51004.shtml. Accessed May 1, 2008.
133. United States Department of Health and Human Services, Embryonic Stem
Cell Research, Fact Sheet. http://www.hhs.gov/news/press/2004pres/
20040714b.html. Accessed 5/1/2008.
T he identification of stem cells in the CNS is the single most impor-
tant scientific discovery in our lifetime. Let me repeat that: the sin-
gle most important scientific discovery in our lifetime!
134. Deleted in proof. This report provides a chronology of the major events in the evolu-
135. Weissman IL: Stem cells: Units of development, units of regeneration, and tion of stem cell work in the CNS. It serves as a reference for all of us
units in evolution. Cell 100:157–168, 2000. to appreciate how our discipline has evolved. The residents that we
136. Wernig M, Meissner A, Foreman R, Brambrink T, Ku M, Hochedlinger K,
train today will look back at this era as a turning point in the treatment
Bernstein BE, Jaenisch R: In vitro reprogramming of fibroblasts into a
pluripotent ES-cell-like state. Nature 448:318–324, 2007.
of neurological disease. The future of stem cell work will alter their per-
137. Westerlund U, Moe MC, Varghese M, Berg-Johnsen J, Ohlsson M, Langmoen ceptions of CNS tumors, vascular disease, degenerative disease, and
IA, Svensson M: Stem cells from the adult human brain develop into func- trauma. This is a wonderful time to enter the field of neurosurgery.
tional neurons in culture. Exp Cell Res 289:378–383, 2003. Current residents will someday use the tools described here to cure the
138. Wilmut I, Schnieke AE, McWhir J, Kind AJ, Campbell KH: Viable offspring diseases and injuries that we can only manage. This report is an intro-
derived from fetal and adult mammalian cells. Nature 385:810–813, 1997. duction into how we arrived at this place. Subsequent reports on stem
139. Wobus AM, Holzhausen H, Jäkel P, Schöneich J: Characterization of a cell biology and research will show our readers where we are headed.
pluripotent stem cell line derived from a mouse embryo. Exp Cell Res
152:212–219, 1984. Joseph M. Piepmeier
140. Wu DC, Boyd AS, Wood KJ: Embryonic stem cell transplantation: Potential New Haven, Connecticut
applicability in cell replacement therapy and regenerative medicine. Front
Biosci 12:4525–4535, 2007.
141. Xie T, Spradling A: Decapentaplegic is essential for the maintenance and divi-
sion of germline stem cells in the Drosophila ovary. Cell 94:251–260, 1998.
142. Xu C: Characterization and evaluation of human embryonic stem cells.
I n this comprehensive treatise, the authors review some of the histor-
ical developments that have led to the current excitement in the
discipline of stem cell biology. From the earliest days, the study of
Methods Enzymol 420:18–37, 2006. embryology and the discovery that stem cells exist have provided
143. Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, Frane JL, Tian S, tremendous promise in terms of potential regeneration/repair of tis-
Nie J, Jonsdottir GA, Ruotti V, Stewart R, Slukvin II, Thomson JA: Induced sues damaged by age and/or disease. The scientists of this discipline
pluripotent stem cell lines derived from human somatic cells. Science
have been very cautious in ensuring that over-interpretation of results
318:1917–1920, 2007.
does not lead to therapeutic adventures that could have disastrous
144. Zwaka TP, Thomson JA: Homologous recombination in human embryonic
stem cells. Nat Biotechnol 21:319–321, 2003. repercussions in terms of unfulfilled outcomes in human patients.
This steady and cautious progress is now beginning to bear fruit: the
most recent discoveries of reprogramming of adult differentiated cells
Acknowledgment back into a stem cell state through re-expression of a set of only 4 genes
We thank James C. Wald, Esq., for his assistance in produing the timeline for (Oct 3/4, Sox2, KLf4, and c-Myc or Oct4, Sox2, NANOG, and Lin28)
this article. provide evidence that only a few master pathways will require re-
instruction to achieve the desired effect of “stemness.” This type of
COMMENTS finding thus opens up avenues of research related to drug or ligand dis-
coveries that may lead one to turn these sets of genes back on, in any

I n this article, Farin et al. provide the first of a 5-part review of stem
cell biology. This component tells the history, from the early work in
the 1960s to dramatic developments in the past few months regarding
cell, to repair a tissue or organ.
The authors provide us with a valuable timeline of the key discover-
ies that have led us to the current state of scientific fervor. The reader will
the production of stem cells from adult mammalian cells. The series is find this a valuable review to bring himself or herself up to date on the
background for neurosurgeons on what, we hope, will be the next nomenclature and key figures of this discipline over the past 50 years.
wave in medicine—and especially in central nervous system (CNS)
regenerative medicine—for battles with cancer, cardiovascular disease, E. Antonio Chiocca
CNS regeneration injuries, or congenital deficits. The authors correctly Columbus, Ohio

34 | VOLUME 64 | NUMBER 1 | JANUARY 2009 www.neurosurgery-online.com


FOUNDATIONS AND LANDMARKS IN STEM CELL BIOLOGY

TABLE A1. Glossarya


Adult (or somatic) stem cell An undifferentiated cell found in a differentiated tissue that can renew itself and differentiate (with certain limitations) to give
rise to all the specialized cell types of the tissue from which it originated; i.e., possessing multipotency and unipotency but not
pluripotency in its native state. Pluripotency has been achieved recently through environmental and genetic manipulations.
Allogeneic transplantation Cell, tissue, or organ transplants from 1 member of a species to a genetically other member of the same species.
Autologous transplantation Cell, tissue, or organ transplants from 1 individual back to the same individual. Such transplants from self do not induce an
immune response and are not rejected. For example, a cancer patient may have her hematopoietic stem cells or bone marrow
removed and stored during treatment with radiation or chemotherapy to kill all blood-forming cells (and, perhaps, all cancer);
her blood-forming capacity is then rescued with autologous hematopoietic stem cells or bone marrow.
Blastocoel The fluid-filled cavity inside the blastocyst of the developing embryo.
Blastocyst A thin-walled hollow sphere made up of an outer layer of cells (trophoblast) that forms the placenta, a fluid filled cavity
(blastocoel), and an inner cell mass containing pluripotent embryonic stem cells. Also called the blastula, the blastocyst is a
preimplantation embryo of about 150 cells produced by cleavage of the zygote after fertilization at approximately 5 days. Further
reproductive development occurs only if the blastocyst is successfully implanted in the uterus.
Blastomere A cell produced during cleavage of a fertilized egg.
Bone marrow stromal cells Also known as mesenchymal stem cells; a mixed population of stem cells derived from the non-blood-forming fraction of bone
marrow that does not give rise to blood cells but instead generates bone, cartilage, fat, and fibrous connective tissue.
Cell culture A technique for growing or maintaining cells on an artificial medium (substance) under laboratory conditions (e.g., Petri dish or
test tube).
Cell division Method by which a single cell divides to create 2 cells. There are 2 main types of cell division: mitosis and meiosis.
Cell line Cells that can be maintained and grown in culture and display an immortal or indefinite life span.
Cell type A specific subset of cells within the body, defined by their appearance, location, and function.
Cell-based therapies Treatment in which stem cells are induced to differentiate into the specific cell type required to repair damaged or destroyed cells
or tissues, alter normal cell response, stimulate native signaling cascades, perform missing metabolic functions, or changing
the normal course of repair into true regeneration.
Cellular reprogramming Transformation of a somatic cell into a pluripotent one via manipulation of regulatory genes and gene products that govern the
state of pluripotency with the purpose of efficiently generating human isogenic pluripotent stem cells and improving the efficiency
of somatic cell nuclear transfer.
Chondrocyte The functional cell type that makes cartilage for joints, ear canals, trachea, epiglottis, larynx, the discs between vertebrae, and
the ends of ribs.
Cleavage The early divisions of the fertilized egg.
Clone To generate identical copies of a molecule, cell, or organism. When it is used to refer to cells grown in a tissue culture dish, a
clone is a line of cells that is genetically identical to the originating cell. This cloned line is produced by cell division (mitosis)
of the originating cell. The term clone may also be used to refer to an animal produced by somatic cell nuclear transfer.
Cloning The process in which an organism, molecule, or cell produces 1 or more genetically alike copies of itself by asexual means.
Cloning may occur by propagation of cuttings, as in the case of plants; continual budding, as in the case of hydra; fission, as in
the case of bacteria and protozoa; parthenogenic asexual reproduction, as in the case of aphids; or somatic cell nuclear transfer,
as in the case of higher-order animals such as mammals. The term cloning can also be applied to a group of cells undergoing replication
by repetitive mitoses (cell divisions). “Therapeutic cloning” creates a line of stem cells genetically identical to the originating cell
for use in research. “Reproductive cloning” creates an organism genetically identical to the organism providing the originating cell.
Culture medium The liquid that covers cells in a culture dish and contains nutrients to feed the cells. Medium may also include other growth factors
added to produce desired changes in the cells.
Deoxyribonucleic acid (DNA) A chemical found primarily in the nucleus of cells; the genetic code.
Differentiation Progressive restriction of the developmental potential and increasing specialization of function which takes place during the
development of the embryo and leads to the formation of specialized cells, tissues, and organs.
Directed differentiation Manipulating stem cell culture conditions to induce differentiation into a particular cell type.
Ectoderm The outermost of 3 germ layers derived from the inner cell mass of the blastocyst that gives rise in later development to the skin,
cells of the amnion and chorion, nervous system, enamel of the teeth, lens of the eye, and neural crest.
Embryo The product of a fertilized egg, from the zygote until the fetal stage.
Embryoid bodies Spheroid colonies seen in culture, produced by the growth of embryonic stem cells in suspension. Embryoid bodies contain
cell types derived from all 3 germ layers, and the distribution and timing of the appearance of specific cell types corresponds to
that observed within the embryo.

NEUROSURGERY VOLUME 64 | NUMBER 1 | JANUARY 2009 | 35


FARIN ET AL.

TABLE A1. continued.


Embryonic germ cells Pluripotent stem cells that are derived from early germ cells that exist between the blastocyst stage of development until their
conversion within gonads to egg or sperm stem cells. Embryonic germ cells are thought to have properties similar to those of embryonic
stem cells.
Embryonic stem cells Pluripotent undifferentiated cell lines established usually from the inner cell mass of the blastocyst stage (5-day-old embryo) of
development. Within the population of cultured embryonic stem cells are cells that can produce more embryonic stem cells or,
under conditions of differentiation, give rise to collections of cells that include most, if not all, cell types that can be found in a
postimplantation embryo, fetus, or developed organism, but not trophoblast or placenta. To date, no embryonic stem cells
cultured in vitro can give rise to developed organisms or even developed organs. A test of pluripotency of mouse embryonic stem
cells is to inject them into blastocysts before implantation; the progeny of these cells can participate in all germline and somatic
tissues along with host blastocyst-derived cells. By definition, an embryonic stem cell is self-renewing (can replicate itself), is pluripotent
(can form all cell types found in the body), and theoretically is immortal.
Endoderm The inner of 3 germ layers of the early embryo that gives rise in later development to tissues such as the lungs, the intestine, the
liver, and the pancreas.
Enucleated A cell with its nucleus removed.
Fertilization The joining of the male gamete (sperm) and the female gamete (egg).
Fetus The stage in development from the end of the embryonic stage, 7–8 weeks after fertilization, to developed organism that ends
at birth.
Fibroblast A connective or support cell found within most tissues of the body, such as skin. Fibroblasts provide an instructive support
scaffold to help the functional cell types of a specific organ perform correctly.
Gamete A reproductive cell containing half of the genetic material necessary to form a complete human organism. During fertilization,
male and female gametes (sperm and ovum, respectively) fuse, producing a zygote.
Gene A functional unit of heredity that is a segment of DNA found on chromosomes in the nucleus of a cell. Genes direct the formation
of an enzyme or other protein.
Germ layers Fertilization of an egg stimulates cell division, and the resulting cells are organized into 3 different layers, called germ layers. The
3 germ layers are the endoderm, mesoderm, and ectoderm and are the 3 precursory tissue layers of the early, primitive embryo
(which form at approximately 2 weeks in the human) that give rise to all tissues of the body.
Germline cells Cells that arise from the inner cell mass and are irreversibly committed to give rise to eggs or sperm. Primitive germline cells (which
are germline stem cells) migrate from the posterior of the early postimplantation embryo to the developing gonads, known as genital
ridges, where they commit to spermatogenesis (in testes) and oogenesis (in ovaries).
Hematopoiesis The development and formation of various types of blood cells.
Hematopoietic cell Transplantation of cells with blood-forming potential, usually bone marrow. Hematopoietic stem cells
transplantation provide rapid and sustained reconstitution of blood formation and are found in adult bone marrow, umbilical cord blood,
mobilized peripheral blood (the nucleated cell fraction of blood after treatment of the donor with agents that increase the passage
of hematopoietic stem cells from bone marrow to blood), and fetal liver.
Hematopoietic Progenitor cells from which all blood cells derive (red blood cells, white blood cells, platelets).
stem cells Hematopoietic stem cells give rise to 2 distinct cells: a replica of the stem cell and a cell that will further proliferate and
differentiate into a mature blood cell. Hematopoietic cells are found within the bone marrow of adults. In the fetus, hematopoietic
cells are found within the liver, spleen, bone marrow, and amniotic fluid surrounding the fetus in the womb. These precursors
of mature blood cells are defined by their ability to replace the bone marrow system after its obliteration (for example, by
[gamma]-irradiation) and can continue to produce mature blood cells.
Hepatocyte The functional cell type of the liver that makes enzymes for detoxifying metabolic waste, destroying red blood cells and reclaiming
their constituents, and synthesis of proteins for blood plasma.
Histocompatible A tissue or organ from a donor (the person giving the organ or tissue) that will not be rejected by the recipient (the patient in whom
the tissue or organ is transplanted). Rejection is caused because the immune system of the recipient sees the transplanted organ
or tissue as foreign and attempts to destroy it. Tissues from 2 random people are rarely histocompatible. In siblings, the probability
of histocompatibility is higher, while identical twins are almost always histocompatible.
Homologous Similar or uniform, often used in the context of genes and DNA sequences. In the context of stem cells, the term homologous
recombination is a technique used to disable a gene in embryonic stem cells.
Homologous A technique used to inactivate a gene and determine its function in a living animal. The process of
recombination homologous recombination is more efficient in embryonic stem cells than in other cell types. It is achieved by introducing a stretch
of DNA that is similar or identical (homologous) to part of a gene and to some of the DNA surrounding the gene, but different
(not homologous) to a specific section of the gene. The DNA is then introduced into the stem cells, and the stretch of homologous
DNA will recognize the similar sequences of the gene within the cell and replace it. The cell is then left with a segment of DNA
in the gene that has the incorrect sequence, which interrupts gene function. The gene is then said to be knocked out. From these
embryonic stem cells, an entire mouse can be made by injecting the altered stem cells into a blastocyst, and implanting the
blastocyst into a female mouse. This is a way to make genetically engineered mice with altered gene function. These experiments
are crucial to understand how specific genes work and interact in living animals.

36 | VOLUME 64 | NUMBER 1 | JANUARY 2009 www.neurosurgery-online.com


FOUNDATIONS AND LANDMARKS IN STEM CELL BIOLOGY

TABLE A1. continued.


Human embryonic A pluripotent stem cell that is derived from the inner cell mass of a blastocyst and can differentiate into several tissue types in a
stem cell dish. They are similar to embryonic stem cells from the mouse; however, human embryonic stem cells are harder to grow than
mouse embryonic stem cells.
In vitro Latin for “in glass”; in a laboratory dish or test tube; an artificial environment.
In vitro fertilization A technique in which an egg is fertilized by sperm outside the body. For use in assisted reproduction, the fertilized egg is
implanted in the uterus at approximately 3 to 4 days of cell division for the purpose of development into a baby. For use in
research, the fertilized egg is maintained in cell culture until it develops into the blastocyst stage at approximately 5 days of
cell division and stem cells can be removed.
Inner cell mass A small group of cells attached to the wall of the blastocyst (the embryo at a very early stage of development). Embryonic stem
cells are made by isolating and culturing the cells that make up the inner cell mass. In development, it is the inner cell mass that
will eventually give rise to all the organs and tissues of the future embryo and fetus, but it does not give rise to the extraembryonic
tissues, such as the placenta.
Long-term self-renewal The ability of stem cells to renew themselves by dividing into the same nonspecialized cell type over long periods (many months
to years) depending on the specific type of stem cell.
Meiosis Cell division of a gamete to reduce the chromosomes within it to half the normal number. This is to ensure that fertilization
restores the full number of chromosomes rather than causing aneuploidy, or an abnormal number of chromosomes.
Mesenchymal stem cells Also known as bone marrow stromal cells; cells from the immature embryonic connective tissue mainly found in bone marrow
that can develop into distinct mesenchymal tissue such as bone, fat, tendons, lymphatic tissue, muscles, adipose tissue, cartilage,
nervous tissue, and blood and blood vessels.
Mesoderm Middle germ layer of a group of cells derived from the inner cell mass of the blastocyst; it gives rise to bone, muscle, connective
tissue, kidneys, and related structures.
Microenvironment The molecules and compounds, such as nutrients and growth factors in the fluid surrounding a cell in an organism or in the laboratory,
that play an important role in determining the characteristics of the cell.
Mitosis Cell division that allows a population of cells to increase its numbers or to maintain its numbers.
Morphology Study of the shape and visual appearance of cells, tissues, and organs.
Morula A globular solid mass of cells (called blastomeres) formed by cleavage of a zygote.
Multipotency Ability of a single stem cell to develop into more than 1 cell type of the body.
Multipotent stem cells Stem cells whose progeny are of multiple differentiated cell types, but all within a particular tissue, organ, or physiological
system. These relatively undifferentiated cells of the same lineage retain the ability to divide and cycle throughout postnatal life
to provide cells that can become specialized and take the place of those that die or are lost. For example, blood-forming
(hematopoietic) stem cells are single multipotent cells that can produce progeny that include hematopoietic stem cells, blood
cell-restricted oligopotent progenitors, and all cell types and elements (e.g., platelets) that are normal components of the blood.
Other examples include myoblasts, myeloid progenitor cells, fibroblasts, tumor stem cells, and skin stem cells.
Myocyte The functional cell type of muscles.
Neural stem cell A type of stem cell that resides in the brain and can make new neurons and glia (oligodendrocytes and astrocytes).
Oligodendrocyte A supporting cell that provides insulation to nerve cells by forming a myelin sheath (a fatty layer) around axons.
Oligopotent progenitor cells Progenitor cells that can produce more than 1 type of mature cells. For example, the clonal common myeloid progenitor is a progenitor
cell which can give rise to blood granulocytes, monocytes, red blood cells, platelets, basophils, eosinophils, and dendritic cells,
but not T lymphocytes, B lymphocytes, or natural killer cells.
Oocyte An egg before maturation; a female gametocyte; also called an ovocyte.
Parthenogenesis A form of reproduction in which an egg is activated without the fusion of sperm with the egg cell. The egg behaves as if it has
been fertilized. Parthenogenesis occurs commonly among insects and other arthropods. Artificially inducing parthenogenesis with
human eggs may be a means to isolate stem cells from an embryo without fertilization.
Passage A round of cell growth and proliferation in cell culture.
Phenotype The description of the characteristics of a cell, a tissue, or an animal. For example, black and white fur of a mouse are 2
phenotypes that can be found. The phenotype is determined by the genes (or the genotype) and by the environment. For example,
short stature is a phenotype that can be genetically determined (and therefore inherited from the parents), but it can also be
caused by malnourishment during childhood (and therefore be caused by the environment).
Placenta The oval spongy structure in the uterus from which the fetus derives its nourishment and oxygen. The placenta develops from
the outer cell layer of the blastocyst, called the trophoblast.
Plasticity The ability of stem cells from an adult tissue to generate the differentiated cell types of another tissue.
Pluripotency Ability of a single stem cell to give rise to all of the various cell types that make up the body. Pluripotent cells cannot make so-
called “extraembryonic” tissues, such as the amnion, chorion, and other components of the placenta. Scientists demonstrate
pluripotency by providing evidence of stable developmental potential, even after prolonged culture, to form derivatives of all 3
embryonic germ layers from the progeny of a single cell and to generate a teratoma after injection into an immunosuppressed
mouse.

NEUROSURGERY VOLUME 64 | NUMBER 1 | JANUARY 2009 | 37


FARIN ET AL.

TABLE A1. continued.


Pluripotent stem cells Stem cells that include in their progeny all cell types that can be found in a postimplantation embryo, fetus, or developed
organism, but not embryonic components of the trophoblast and placenta (these are usually called extraembryonic). Pluripotent
stem cells give rise to multipotent and unipotent stem cells as the embryo develops.
Polar body Structure produced when an early egg cell, or oogonium, undergoes meiosis. In the first meiosis, the oogonium divides its
chromosomes evenly between the 2 cells but divides its cytoplasm unequally. One cell retains most of the cytoplasm, while the
other gets almost none, leaving it very small. This smaller cell is called the first polar body. The first polar body usually degenerates.
The ovum, or larger cell, then divides again, producing a second polar body with half the amount of chromosomes but almost no
cytoplasm. The second polar body splits off and remains adjacent to the large cell, or oocyte, until it (the second polar body)
degenerates. Only 1 large functional oocyte, or egg, is produced at the end of meiosis.
Postimplantation embryos Implanted embryos in all early stages of development until the establishment of the body plan of a developed organism with identifiable
tissues and organs.
Pre-embryos Eggs that are fertilized and develop to approximately 2–3 days old in a laboratory (in vitro fertilization) and are ready for
implantation for the purposes of human reproduction or removal of stem cells for the purposes of research.
Preimplantation With regard to an embryo, preimplantation indicates that the embryo has not yet implanted in the wall of the uterus. Human embryonic
stem cells are derived from preimplantation stage embryos fertilized outside a woman’s body (in vitro).
Preimplantation embryos Fertilized eggs (zygotes) and all of the developmental stages up to, but not beyond, the blastocyst stage.
Primitive streak The beginning of the vertebral column in the human embryo that develops at approximately 14 days after conception.
Progenitor cell Derived from stem cells and intermediate to the production of mature cells. Often incorrectly confused with stem cell; an early
descendant of a stem cell that can only differentiate, but not renew itself. In contrast, a stem cell exhibits potency and self-
renewal. A progenitor cell is often more limited in the types of cells it may develop into, compared with a stem cell, i.e., it
exhibits increasing differentiation.
Proliferation Expansion of cells by the continuous division of single cells into 2 identical daughter cells.
Regenerative medicine A treatment in which stem cells are induced to differentiate into the specific cell type required to repair damaged or destroyed
cell populations or tissues. (See also Cell-based therapies.)
Reproductive cloning Creation of an animal being identical to the animal that donated the somatic cell nucleus. The embryo is implanted in a uterus
and develops into a live being. The first animal to be created by reproductive cloning was Dolly, the sheep born at the Roslin
Institute in Scotland in 1996. (See also Somatic cell nuclear transfer).
Signals Internal and external factors that control changes in cell structure and function.
Somatic cell nuclear transfer A technique that combines an enucleated egg (nucleus removed) and the nucleus of a somatic cell to make an embryo. Somatic
cell nuclear transfer can be used for therapeutic or reproductive purposes, but the initial stage that combines an enucleated egg
and a somatic cell nucleus is the same. With regard to reproductive cloning, somatic cell nuclear transfer attempts to produce
a fetus that is implanted into a woman’s uterus; the live offspring is genetically identical to the donor of the somatic cell DNA.
Somatic cells All cells within the developing or developed organism with the exception of germline cells.
Somatic stem cells Nonembryonic stem cells that are not derived from gametes (egg or sperm cells).
Stem cell line Stem cells that have been growing in cell culture for 6 or more months without becoming specialized and appear genetically normal.
Stem cell transplantation The transfer of stem cells from 1 individual to another within the same species (homologous transplantation) or between species
(xenotransplantation), or transfer within the same individual (autologous transplantation). The source and location of the stem
cells determines their potency or pluripotency to differentiate into various cell types.
Stem cells Cells with the ability to divide for indefinite periods in culture (self-renewal) and to give rise to specialized cells (potency).
Stromal cells Non-blood cells derived from blood organs, such as bone marrow or fetal liver, which are capable of supporting growth of
blood cells in vitro. Stromal cells that make the matrix within the bone marrow are derived from mesenchymal stem cells.
Subculturing Transferring cultured cells, with or without dilution, from 1 culture vessel to another.
Surface markers Proteins on the outside surface of a cell that are unique to certain cell types, which are visualized using antibodies or other
detection methods.
Teratoma Scientists verify that they have established a human embryonic stem cell line by injecting putative stem cells into mice with a
dysfunctional immune system. Since the injected cells cannot be destroyed by the mouse’s immune system, they survive and form
a multilayered benign tumor called a teratoma. Teratomas serve to establish the ability of a stem cell to give rise to all cell types
in the body, as teratomas contain cells derived from each of the 3 embryonic germ layers.
Therapeutic cloning Somatic cell nuclear transfer, or cloning, for the isolation of embryonic stem cells that exactly match an individual. The embryonic
stem cells are derived from the blastocyst (before it becomes a fetus) and can be instructed to form particular cell types (e.g., heart
muscle) to be implanted into damaged tissue (e.g., heart) to restore its function. If the stem cells are placed back into the
individual who donated the DNA for the somatic cell nuclear transfer, the embryonic stem cells and their derivatives are
genetically identical and thus immunocompatible (they will not be rejected).

38 | VOLUME 64 | NUMBER 1 | JANUARY 2009 www.neurosurgery-online.com


FOUNDATIONS AND LANDMARKS IN STEM CELL BIOLOGY

TABLE A1. continued.


Totipotent stem cells Stem cells that include in their progeny all cell types that can be found in an embryo, fetus, or developed organism, as well as
the cells that form the embryonic components of the trophoblast and placenta required to support development and birth.
Considered the “master cells of the body.” The zygote and the cells at the very early stages after fertilization (i.e., the 2-cell
stage) are considered totipotent.
Transdifferentiation The ability of a particular cell of 1 tissue, organ, or system, including stem or progenitor cells, to be caused to differentiate into
a cell type characteristic of another tissue, organ, or system; e.g., hematopoietic stem cells to liver hepatocytes.
Transplantation biology The science that studies the transplantation of organs and cells, include graft rejection.
Trophoblast The extraembryonic tissue (outer layer of cells of the blastocyst) responsible for uterine implantation, developing into the placenta,
and controlling the exchange of oxygen and metabolites between mother and embryo. In contrast to embryonic stem cells, the
trophoblast does not come from the inner cell mass, but from cells surrounding it.
Umbilical cord blood Hematopoietic stem cells collected from the umbilical cord during and shortly after delivery. These stem cells are in the
stem cells blood at the time of delivery because they transition from the liver, where blood formation takes place during fetal life, to the
bone marrow, where blood is made postnatally. Umbilical cord blood stem cells are similar to stem cells that reside in bone marrow,
and they can be used for the treatment of leukemia and other diseases of the blood. Cord blood is currently used to rescue
patients who have undergone chemotherapy to destroy their bone marrow. Efforts are now being undertaken to consistently
harvest and store these cells for future potential use.
Undifferentiated cell A cell that has not yet generated structures or manufactured proteins characteristic of a specialized cell type.
Unipotent stem cells Stem cells that self-renew as well as give rise to a single mature cell type; e.g., spermatogenic stem cells.
Wharton’s jelly A gelatinous substance within the umbilical cord; it has recently been shown to be a source of potentially pluripotent stem cells.
Zygote The cell formed by the union of 2 gametes; the result of sperm-egg interactions leading to the fusion of sperm and egg nuclei.
The zygote usually begins cell division to give rise to the stages of the preimplantation embryo, from the 2-cell stage to the
blastocyst; however, only the fertilized egg is the zygote.

a
Sources: National Institutes of Health, International Society for Stem Cell Research, Cambridge Healthtech Institute, and the University of Kansas.

Mario R. Capecchi (left), Sir Martin J. Evans (center), and Oliver Smithies (right), recipients of the Nobel Prize in Physiology or Medicine in 2007 “for
their discoveries of principles for introducing specific gene modifications in mice by the use of embryonic stem cells.” See Apuzzo, p 1, and Farin et al.,
pp 15–39.

NEUROSURGERY VOLUME 64 | NUMBER 1 | JANUARY 2009 | 39

You might also like