Cellular Differentiation: Mammalian Cell Types

Download as docx, pdf, or txt
Download as docx, pdf, or txt
You are on page 1of 7

Cellular differentiation

From Wikipedia, the free encyclopedia


"Cell differentiation" redirects here. For the journal, see Cell Differentiation (journal).
In developmental biology, cellular differentiation is the process by which a less
specialized cell becomes a more specialized cell type. Differentiation occurs numerous times
during the development of amulticellular organism as the organism changes from a
simple zygote to a complex system of tissues and cell types. Differentiation continues in
adulthood as adult stem cells divide and create fully differentiated daughter cells during tissue
repair and during normal cell turnover. Differentiation dramatically changes a cell's size,
shape, membrane potential, metabolic activity, and responsiveness to signals. These changes
are largely due to highly controlled modifications in gene expression. With a few exceptions,
cellular differentiation almost never involves a change in the DNA sequence itself. Thus, different
cells can have very different physical characteristics despite having the same genome.
A cell that can differentiate into all cell types of the adult organism is known as pluripotent. Such
cells are called embryonic stem cells in animals and meristematic cells in higher plants. A cell
that can differentiate into all cell types, including the placental tissue, is known as totipotent. In
mammals, only the zygote and subsequent blastomeres are totipotent, while in plants many
differentiated cells can become totipotent with simple laboratory techniques. In cytopathology,
the level of cellular differentiation is used as a measure of cancer progression. "Grade" is a
marker of how differentiated a cell in a tumor is.[1]

Contents
  [hide] 

 1 Mammalian cell types


 2 Dedifferentiation
 3 Mechanisms
o 3.1 Epigenetic control of cellular differentiation
 3.1.1 Importance of epigenetic control
 3.1.2 Mechanisms of epigenetic regulation
 3.1.2.1 Pioneering factors (Oct4, Sox2, Nanog)
 3.1.2.2 Polycomb repressive complex (PRC2)
 3.1.2.3 Trithorax group proteins (TrxG)
 3.1.2.4 DNA methylation
 3.1.2.5 Nucleosome positioning
 3.1.3 Role of signaling in epigenetic control
 4 See also
 5 References

Mammalian cell types[edit]


See also: List of distinct cell types in the adult human body
Three basic categories of cells make up the mammalian body: germ cells, somatic cells,
and stem cells. Each of the approximately 100 trillion (1014) cells in an adult human has its own
copy or copies of the genome except certain cell types, such as red blood cells, that lack nuclei
in their fully differentiated state. Most cells are diploid; they have two copies of
each chromosome. Such cells, called somatic cells, make up most of the human body, such as
skin and muscle cells. Cells differentiate to specialize for different functions. [citation needed]
Germ line cells are any line of cells that give rise to gametes—eggs and sperm—and thus are
continuous through the generations. Stem cells, on the other hand, have the ability to divide for
indefinite periods and to give rise to specialized cells. They are best described in the context of
normal human development.[citation needed]
Development begins when a sperm fertilizes an egg and creates a single cell that has the
potential to form an entire organism. In the first hours after fertilization, this cell divides into
identical cells. In humans, approximately four days after fertilization and after several cycles of
cell division, these cells begin to specialize, forming a hollow sphere of cells, called a blastocyst.
[2]
 The blastocyst has an outer layer of cells, and inside this hollow sphere, there is a cluster of
cells called the inner cell mass. The cells of the inner cell mass go on to form virtually all of the
tissues of the human body. Although the cells of the inner cell mass can form virtually every type
of cell found in the human body, they cannot form an organism. These cells are referred to
as pluripotent.[citation needed]
Pluripotent stem cells undergo further specialization into multipotent progenitor cells that then
give rise to functional cells. Examples of stem and progenitor cells include: [citation needed]

 Hematopoietic stem cells (adult stem cells) from the bone marrow that give rise to red
blood cells, white blood cells, and platelets
 Mesenchymal stem cells (adult stem cells) from the bone marrow that give rise to stromal
cells, fat cells, and types of bone cells
 Epithelial stem cells (progenitor cells) that give rise to the various types of skin cells
 Muscle satellite cells (progenitor cells) that contribute to differentiated muscle tissue.
A pathway that is guided by the cell adhesion molecules consisting of four amino
acids, arginine, glycine, asparagine, and serine, is created as the cellular
blastomere differentiates from the single-layered blastula to the three primary layers of germ
cells in mammals, namely the ectoderm, mesoderm and endoderm (listed from most distal
(exterior) to proximal (interior)). The ectoderm ends up forming the skin and the nervous system,
the mesoderm forms the bones and muscular tissue, and the endoderm forms the internal organ
tissues.

Dedifferentiation[edit]

Micrograph of a liposarcoma with some dedifferentiation, that is not identifiable as a liposarcoma, (left edge of
image) and a differentiated component (with lipoblasts and increased vascularity (right of image)). Fully
differentiated (morphologically benign) adipose tissue (center of the image) has few blood vessels. H&E stain.

Dedifferentiation is a cellular process often seen in more basal life forms such


as worms and amphibians in which a partially or terminally differentiated cell reverts to an earlier
developmental stage, usually as part of a regenerative process.[3][4] Dedifferentiation also occurs
in plants.[5] Cells in cell culture can lose properties they originally had, such as protein expression,
or change shape. This process is also termed dedifferentiation. [6]
Some believe dedifferentiation is an aberration of the normal development cycle that results
in cancer,[7] whereas others believe it to be a natural part of the immune response lost by humans
at some point as a result of evolution.
A small molecule dubbed reversine, a purine analog, has been discovered that has proven to
induce dedifferentiation in myotubes. These dedifferentiated cells could then redifferentiate
into osteoblasts and adipocytes.[8]
Dedifferentiation to totipotency or pluripotency: an overview of methods.'Various methods exist to revert adult
somatic cells to pluripotency or totipotency. In the case of totipotency, reprogramming is mediated through a
mature metaphase II oocyte as in somatic cell nuclear transfer (Wilmut et al., 1997). Recent work has
demonstrated the feasibility of enucleated zygotes or early blastomeres chemically arrested during mitosis, such
that nuclear envelope break down occurs, to support reprogramming to totipotency in a process called
chromosome transfer (Egli and Eggan, 2010). Direct reprogramming methods support reversion to pluripotency;
though, vehicles and biotypes vary considerably in efficiencies (Takahashi and Yamanaka, 2006). Viral-mediated
transduction robustly supports dedifferentiation to pluripotency through retroviral or DNA-viral routes but carries
the onus of insertional inactivation. Additionally, epigenetic reprogramming by enforced expression of OSKM
through DNA routes exists such as plasmid DNA, minicircles, transposons, episomes and DNA mulicistronic
construct targeting by homologous recombination has also been demonstrated; however, these methods suffer
from the burden to potentially alter the recipient genome by gene insertion (Ho et al., 2010). While protein-
mediated transduction supports reprogramming adult cells to pluripotency, the method is cumbersome and
requires recombinant protein expression and purification expertise, and reprograms albeit at very low frequencies
(Kim et al., 2009). A major obstacle of using RNA for reprogramming is its lability and that single-stranded RNA
biotypes trigger innate antiviral defense pathways such as interferon and NF-κB-dependent pathways. In vitro
transcribed RNA, containing stabilizing modifications such as 5-methylguanosine capping or substituted
ribonucleobases, e.g. pseudouracil, is 35-fold more efficient than viral transduction and has the additional benefit
of not altering the somatic genome (Warren et al., 2010).

Mechanisms[edit]

Mechanisms of cellular differentiation

Each specialized cell type in an organism expresses a subset of all the genes that constitute


the genome of that species. Each cell type is defined by its particular pattern of regulated gene
expression. Cell differentiation is thus a transition of a cell from one cell type to another and it
involves a switch from one pattern of gene expression to another. Cellular differentiation during
development can be understood as the result of a gene regulatory network. A regulatory gene
and its cis-regulatory modules are nodes in a gene regulatory network; they receive input and
create output elsewhere in the network.[9] The systems biology approach to developmental
biology emphasizes the importance of investigating how developmental mechanisms interact to
produce predictable patterns (morphogenesis). (However, an alternative view has been proposed
recently. Based on stochastic gene expression, cellular differentiation is the result of a Darwinian
selective process occurring among cells. In this frame, protein and gene networks are the result
of cellular processes and not their cause. See: Cellular Darwinism)
A few evolutionarily conserved types of molecular processes are often involved in the cellular
mechanisms that control these switches. The major types of molecular processes that control
cellular differentiation involve cell signaling. Many of the signal molecules that convey information
from cell to cell during the control of cellular differentiation are called growth factors. Although the
details of specific signal transduction pathways vary, these pathways often share the following
general steps. A ligand produced by one cell binds to a receptor in the extracellular region of
another cell, inducing a conformational change in the receptor. The shape of the cytoplasmic
domain of the receptor changes, and the receptor acquires enzymatic activity. The receptor then
catalyzes reactions that phosphorylate other proteins, activating them. A cascade of
phosphorylation reactions eventually activates a dormant transcription factor or cytoskeletal
protein, thus contributing to the differentiation process in the target cell. [10] Cells and tissues can
vary in competence, their ability to respond to external signals. [11]
Signal induction refers to cascades of signaling events, during which a cell or tissue signals to
another cell or tissue to influence its developmental fate. [11] Yamamoto and Jeffery[12] investigated
the role of the lens in eye formation in cave- and surface-dwelling fish, a striking example of
induction.[11] Through reciprocal transplants, Yamamoto and Jeffery[12] found that the lens vesicle
of surface fish can induce other parts of the eye to develop in cave- and surface-dwelling fish,
while the lens vesicle of the cave-dwelling fish cannot. [11]
Other important mechanisms fall under the category of asymmetric cell divisions, divisions that
give rise to daughter cells with distinct developmental fates. Asymmetric cell divisions can occur
because of asymmetrically expressed maternal cytoplasmic determinants or because of
signaling.[11] In the former mechanism, distinct daughter cells are created
during cytokinesis because of an uneven distribution of regulatory molecules in the parent cell;
the distinct cytoplasm that each daughter cell inherits results in a distinct pattern of differentiation
for each daughter cell. A well-studied example of pattern formation by asymmetric divisions
is body axis patterning in Drosophila. RNA molecules are an important type of intracellular
differentiation control signal. The molecular and genetic basis of asymmetric cell divisions has
also been studied in green algae of the genus Volvox, a model system for studying how
unicellular organisms can evolve into multicellular organisms. [11]In Volvox carteri, the 16 cells in
the anterior hemisphere of a 32-cell embryo divide asymmetrically, each producing one large and
one small daughter cell. The size of the cell at the end of all cell divisions determines whether it
becomes a specialized germ or somatic cell.[11][13]
Epigenetic control of cellular differentiation [edit]
Main article: Epigenetics in stem cell differentiation
Since each cell, regardless of cell type, possesses the same genome, determination of cell type
must occur at the level of gene expression. While the regulation of gene expression can occur
through cis- and trans-regulatory elements including a gene’s promoter and enhancers, the
problem arises as to how this expression pattern is maintained over numerous generations of cell
division. As it turns out, epigenetic processes play a crucial role in regulating the decision to
adopt a stem, progenitor, or mature cell fate. This section will focus primarily on mammalian stem
cells.
Importance of epigenetic control[edit]
The first question that can be asked is the extent and complexity of the role of epigenetic
processes in the determination of cell fate. A clear answer to this question can be seen in the
2011 paper by Lister R, et al. [14] on aberrant epigenomic programming in human induced
pluripotent stem cells. As induced pluripotent stem cells (iPSCs) are thought to mimic embryonic
stem cells in their pluripotent properties, few epigenetic differences should exist between them.
To test this prediction, the authors conducted whole-genome profiling of DNA
methylation patterns in several human embryonic stem cell (ESC), iPSC, and progenitor cell
lines.
Female adipose cells, lung fibroblasts, and foreskin fibroblasts were reprogrammed into induced
pluripotent state with the OCT4, SOX2, KLF4, and MYC genes. Patterns of DNA methylation in
ESCs, iPSCs, somatic cells were compared. Lister R, et al. observed significant resemblance in
methylation levels between embryonic and induced pluripotent cells. Around 80% of CG
dinucleotides in ESCs and iPSCs were methylated, the same was true of only 60% of CG
dinucleotides in somatic cells. In addition, somatic cells possessed minimal levels of cytosine
methylation in non-CG dinucleotides, while induced pluripotent cells possessed similar levels of
methylation as embryonic stem cells, between 0.5 and 1.5%. Thus, consistent with their
respective transcriptional activities,[14] DNA methylation patterns, at least on the genomic level,
are similar between ESCs and iPSCs.
However, upon examining methylation patterns more closely, the authors discovered 1175
regions of differential CG dinucleotide methylation between at least one ES or iPS cell line. By
comparing these regions of differential methylation with regions of cytosine methylation in the
original somatic cells, 44-49% of differentially methylated regions reflected methylation patterns
of the respective progenitor somatic cells, while 51-56% of these regions were dissimilar to both
the progenitor and embryonic cell lines. In vitro-induced differentiation of iPSC lines saw
transmission of 88% and 46% of hyper and hypo-methylated differentially methylated regions,
respectively.
Two conclusions are readily apparent from this study. First, epigenetic processes are heavily
involved in cell fate determination, as seen from the similar levels of cytosine methylation
between induced pluripotent and embryonic stem cells, consistent with their respective patterns
of transcription. Second, the mechanisms of de-differentiation (and by extension, differentiation)
are very complex and cannot be easily duplicated, as seen by the significant number of
differentially methylated regions between ES and iPS cell lines. Now that these two points have
been established, we can examine some of the epigenetic mechanisms that are thought to
regulate cellular differentiation.
Mechanisms of epigenetic regulation[edit]
Pioneering factors (Oct4, Sox2, Nanog)[edit]
Three transcription factors, OCT4, SOX2, and NANOG – the first two of which are used in iPSC
reprogramming – are highly expressed in undifferentiated embryonic stem cells and are
necessary for the maintenance of their pluripotency.[15] It is thought that they achieve this through
alterations in chromatin structure, such as histone modification and DNA methylation, to restrict
or permit the transcription of target genes.
Polycomb repressive complex (PRC2)[edit]
In the realm of gene silencing, Polycomb repressive complex 2, one of two classes of
the Polycomb group (PcG) family of proteins, catalyzes the di- and tri-methylation of histone H3
lysine 27 (H3K27me2/me3).[16][15][17] By binding to the H3K27me2/3-tagged nucleosome, PRC1
(also a complex of PcG family proteins) catalyzes the mono-ubiquitinylation of histone H2A at
lysine 119 (H2AK119Ub1), blocking RNA polymerase II activity and resulting in transcriptional
suppression.[15] PcG knockout ES cells do not differentiate efficiently into the three germ layers,
and deletion of the PRC1 and PRC2 genes leads to increased expression of lineage-affiliated
genes and unscheduled differentiation. [15] Presumably, PcG complexes are responsible for
transcriptionally repressing differentiation and development-promoting genes.
Trithorax group proteins (TrxG)[edit]
Alternately, upon receiving differentiation signals, PcG proteins are recruited to promoters of
pluripotency transcription factors. PcG-deficient ES cells can begin differentiation but cannot
maintain the differentiated phenotype. [15] Simultaneously, differentiation and development-
promoting genes are activated by Trithorax group (TrxG) chromatin regulators and lose their
repression.[15][17] TrxG proteins are recruited at regions of high transcriptional activity, where they
catalyze the trimethylation of histone H3 lysine 4 (H3K4me3) and promote gene activation
through histone acetylation. [17] PcG and TrxG complexes engage in direct competition and are
thought to be functionally antagonistic, creating at differentiation and development-promoting loci
what is termed a “bivalent domain” and rendering these genes sensitive to rapid induction or
repression.[18]
DNA methylation[edit]
Regulation of gene expression is further achieved through DNA methylation, in which the DNA
methyltransferase-mediated methylation of cytosine residues in CpG dinucleotides maintains
heritable repression by controlling DNA accessibility. [18] The majority of CpG sites in embryonic
stem cells are unmethylated and appear to be associated with H3K4me3-carrying nucleosomes.
[15]
 Upon differentiation, a small number of genes, including OCT4 and NANOG, [18] are methylated
and their promoters repressed to prevent their further expression. Consistently, DNA
methylation-deficient embryonic stem cells rapidly enter apoptosis upon in vitro differentiation.[15]
Nucleosome positioning[edit]
While the DNA sequence of almost all cells of an organism is the same, the binding patterns of
transcription factors and the corresponding gene expression patterns are different. To a large
extent, differences in transcription factor binding are determined by the chromatin accessibility of
their binding sites through histone modification and/or pioneer factors. In particular, it is important
to know whether a nucleosome is covering a given genomic binding site or not. Recent studies
have elucidated the role of nucleosome positioning during stem cell development. [19]
Role of signaling in epigenetic control[edit]
A final question to ask concerns the role of cell signaling in influencing the epigenetic processes
governing differentiation. Such a role should exist, as it would be reasonable to think that
extrinsic signaling can lead to epigenetic remodeling, just as it can lead to changes in gene
expression through the activation or repression of different transcription factors. Interestingly,
little direct data is available concerning the specific signals that influence the epigenome, and the
majority of current knowledge consist of speculations on plausible candidate regulators of
epigenetic remodeling.[20] We will first discuss several major candidates thought to be involved in
the induction and maintenance of both embryonic stem cells and their differentiated progeny, and
then turn to one example of specific signaling pathways in which more direct evidence exists for
its role in epigenetic change.
The first major candidate is Wnt signaling pathway. The Wnt pathway is involved in all stages of
differentiation, and the ligand Wnt3a can substitute for the overexpression of c-Myc in the
generation of induced pluripotent stem cells.[20] On the other hand, disruption of ß-catenin, a
component of the Wnt signaling pathway, leads to decreased proliferation of neural progenitors.
Growth factors comprise the second major set of candidates of epigenetic regulators of cellular
differentiation. These morphogens are crucial for development, and include bone morphogenetic
proteins,transforming growth factors (TGFs), and fibroblast growth factors (FGFs). TGFs and
FGFs have been shown to sustain expression of OCT4, SOX2, and NANOG by downstream
signaling to Smadproteins.[20] Depletion of growth factors promotes the differentiation of ESCs,
while genes with bivalent chromatin can become either more restrictive or permissive in their
transcription.[20]
Several other signaling pathways are also considered to be primary candidates.
Cytokine leukemia inhibitory factors are associated with the maintenance of mouse ESCs in an
undifferentiated state. This is achieved through its activation of the Jak-STAT3 pathway, which
has been shown to be necessary and sufficient towards maintaining mouse ESC pluripotency.
[21]
 Retinoic acid can induce differentiation of human and mouse ESCs, [20] and Notch signaling is
involved in the proliferation and self-renewal of stem cells. Finally, Sonic hedgehog, in addition to
its role as a morphogen, promotes embryonic stem cell differentiation and the self-renewal of
somatic stem cells.[20]
The problem, of course, is that the candidacy of these signaling pathways was inferred primarily
on the basis of their role in development and cellular differentiation. While epigenetic regulation is
necessary for driving cellular differentiation, they are certainly not sufficient for this process.
Direct modulation of gene expression through modification of transcription factors plays a key
role that must be distinguished from heritable epigenetic changes that can persist even in the
absence of the original environmental signals. Only a few examples of signaling pathways
leading to epigenetic changes that alter cell fate currently exist, and we will focus on one of them.
Expression of Shh (Sonic hedgehog) upregulates the production of Bmi1, a component of the
PcG complex that recognizes H3K27me3. This occurs in a Gli-dependent manner,
as Gli1 and Gli2 are downstream effectors of the Hedgehog signaling pathway. In culture, Bmi1
mediates the Hedgehog pathway’s ability to promote human mammary stem cell self-renewal.
[22]
 In both humans and mice, researchers showed Bmi1 to be highly expressed in proliferating
immature cerebellar granule cell precursors. When Bmi1 was knocked out in mice, impaired
cerebellar development resulted, leading to significant reductions in postnatal brain mass along
with abnormalities in motor control and behavior. [23] A separate study showed a significant
decrease in neural stem cell proliferation along with increased astrocyte proliferation in Bmi null
mice.[24]
In summary, the role of signaling in the epigenetic control of cell fate in mammals is largely
unknown, but distinct examples exist that indicate the likely existence of further such
mechanisms.

You might also like