SlideShare una empresa de Scribd logo
1 de 196
Descargar para leer sin conexión
Restorative neurology
                          Essay
                In Neuropsychiatry
   Submitted for partial fulfillment of Master Degree

                           By
           Samy moussa Seliem
                       M.B.B.CH

               Supervisors of


Prof. Mohammed Yasser Metwally
          Professor of Neuropsychiatry

   Faculty of Medicine-Ain Shams University

            www.yassermetwally.com



Prof. Naglaa Mohamed Elkhayat
          Professor of Neuropsychiatry

   Faculty of Medicine-Ain Shams University



   Dr. Haitham Hamdy salem
           Lecturer of Neuropsychiatry

   Faculty of Medicine-Ain Shams University




                     Faculty of Medicine


                    Ain Shams University


                           2011

                                  i
Contents

Subject

1. Introduction and aim of the work
2. Stem cell
3. Stem cell therapy in Parkinson disease
4. Stem cell therapy in in stroke
5. Stem cell therapy in demyelinating disease
6. Stem cell therapy in in amyotrophic lateral sclerosis

7. Stem cell therapy in muscular dystrophy

8. Stem cell therapy Huntington chorea

9. Stem cell therapy for Alzheimer's Disease

10.Stem cell therapy in degenerative diseases in children

11.Stem cell therapy in retinal degeneration

12.Stem cell therapy in spinal cord injury

13.Stem cell therapy in peripheral nerve injury

Summary

References



                                    i
Introduction
   Stem cells are unspecialized cells in the human body that are
capable of becoming specialized cells, each with new
specialized cell functions. The best example of a stem cell is the
bone marrow stem cell that is unspecialized and able to
specialize into blood cells, such as white blood cells and red
blood cells, and these new cell types have special functions,
such as being able to produce antibodies, act as scavengers to
combat infection and transport gases. Thus one cell type stems
from the other and hence the term “stem cell.” Basically, a stem
cell remains uncommitted until it receives a signal to develop
into a specialized cell. Stem cells have the remarkable properties
of developing into a variety of cell types in the human body.
They serve as a repair system by being able to divide without
limit to replenish other cells. When stem cell divides, each new
cell has the potential to either remain as a stem cell or become
another cell type with new special functions, such as blood cells,
brain cells, etc. (Bongso and Lee, 2005).

   Stem cells also known as progenitor cells which are cells
that have not undergone differentiation to acquire specific
structure or role. They have the potential to self-renew, divide
and differentiate into specialized cell types. They are also,
sometimes, termed ‘pluripotent’ or ‘undifferentiated’ cells



                                i
because they can differentiate and develop into various cell lines
(Metwally, 2009).

   Scientists and researchers are interested in stem cells for
several reasons. Although stem cells do not serve any one
function, many have the capacity to serve any function after
they are instructed to specialize. Every cell in the body, for
example, is derived from first few stem cells formed in the early
stages of embryological development. Therefore, stem cells
extracted from embryos can be induced to become any desired
cell type. This property makes stem cells powerful enough to
regenerate damaged tissue under the right conditions (Crosta,
2010).

   Perhaps, the most       important reason that stem cell
development is so appealing to neurologists can be found in the
statement “The adult human brain, in contrast to other organs
such as skin and liver, lacked the capacity for self repair and
regeneration” (Lin et al., 2007).

   The types of stem cells include: Bone marrow-derived
mesenchymal stem cells (BMSCs), embryonic stem cells
(ESCs), Adult (somatic) stem cells, and neural stem cells
(NSCs). BMSCs also termed bone marrow stromal cells are
another example of a somatic stem cell being studied for its
therapeutic potential in the central nervous system (CNS) and in
other tissue (Abdallah and Kassem, 2008).


                                ii
BMSCS generate neurotransmitter-responsive cells with
electro-physiological properties similar to neurons (Diana and
Gabriel, 2008).

   ESCs are pluripotent cells isolated from the inner cell mass
of day 5-8 blastocyte with indefinite self-renewal capabilities as
well as of the ability to differentiate into all cell types derived
from the three embryonic germ layers. The primary therapeutic
goal of ESCs research is cell replacement therapy (Aoki et al.,
2007).

   Adult (somatic) stem cells: it has a capacity to differentiate
into tissue-specific types and represent a potential source of
autologus cells for transplantation therapy that eliminate
immunological complications associated with allogenic donor
cells as well as bypass ethical concern associated with ESCs,
All types are generally characterized by their potency, or
potential to differentiate into different cell types (such as skin,
muscle, bone, etc) (Lin et al., 2007).

   Scientists discovered ways to obtain or derive stem cells
from early mouse embryos more than 20 years ago. Many years
of detailed study of biology of mouse stem cells led to the
discovery, in 1998, of how to isolate stem cells from human
embryos and grow the cells in the laboratory. These are called
human embryonic stem cells. The embryos used in these studies
were created for infertility purposes through in vitro fertilization



                                 iii
procedures and when they were no longer needed for that
purpose, they were donated for research with the informed
consent of the donor (Ordrico et al., 2001).

   The concept that the adult mammalian CNS contains NSCs
was first discovered from evidence of neuronal turnover in the
olfactory bulb and hippocampus in the adult organism cells with
more restricted neural differentiation capabilities committed to
specific subpopulation lineage, have been generated from
human ESCs or directly isolated from neurogenic regions of
fetal and adult CNS, such as the subventricular zone, which
provides neuroblasts to replenish inhibitory interneurons in the
olfactory bulb (Lin et al., 2007).

   Stem cell differentiation must be turned on, given direction,
and turned off as needed in order to properly supply the basic
building blocks of tissues in different organ systems. This
requirement for precise regulation applies to an even greater
degree to the differentiation of neuronal progenitor cells,
because effective neural function depends on establishing
precise linkage and interactions between different individual
neurons and classes of neurons (Metwally, 2009).

   Most tissue repair events in mammals are dedifferentiation
independent events brought about by the activation of pre-
existing stem cells or progenitor cells. By definition, a
progenitor cell lies in between a stem cell and a terminally
differentiated cell (Crosta, 2010).


                                iv
With    the    therapeutic   application   of    NSCs     for
neurorestoration in mind, a clearer picture is emerging. Both in
normal neurodevelopment and stem cell biology, the precursor
cells display preprogrammed behavior modified by cues from
the local environment. The fundamental assumption is that
differentiation and predictable behavior of NSCs can be
achieved if the appropriate cocktail of soluble/diffusible or
contact-mediated signals is present. In addition, several
corollary considerations are quickly evident. For example, can
we use NSCs from different sources in an equivalent fashion?
The answer to this important question requires that we
understand the developmental potential of all the types of NSCs
(Marquez et al., 2005).

   Medical researchers believe that stem cell therapy has the
potential to dramatically change the treatment of human disease.
A number of adult stem cell therapies already exist, particularly
bone marrow transplants that are used to treat leukemia. In the
future, medical researchers anticipate being able to use
technologies derived from stem cell research to treat a wider
variety of diseases including cancer, Parkinson's disease, spinal
cord injuries, Amyotrophic lateral sclerosis, multiple sclerosis,
and muscle damage, amongst a number of other impairments
and conditions (Goldman and Windrem, 2006).




                                v
Aim of the work:

   The aim of this work is to study and summarize recent
progress in stem cell therapies aimed at neurodegenerative
disorder and illustrate how some of aforementioned methods
and strategies are being utilized to formulate clinically viable
treatments.




                              vi
Stem cells

Definition: A stem cell is a cell that has the ability to divide
(self replicate) for indefinite periods, often throughout the life of
the organism. Under the right conditions, or given the right
signals, stem cells can give rise (differentiate) to the many
different cell types that make up the organism. That is, stem
cells have the potential to develop into mature cells that have
characteristic shapes and specialized functions, such as heart
cells, skin cells, or nerve cells (Charron et al., 2009).
   The word “stem” actually originated from old botanical
monographs from the same terminology as the stems of plants,
where stem cells were demonstrated in the apical root and shoot
meristems    that   were    responsible    for   the   regenerative
competence of plants. Hence also the use of word “stem” in
“meristem” (Kiessling and Anderson, 2003).



Historical overview of stem cell therapy:
   The stem cell is the origin of life. As stated first by the great
pathologist (Rudolph Virchow), “All cells come from cells”.
The fertilized egg is formed from fusion of the haploid progeny
of germinal stem cells. The fertilized egg is totipotent; from it
forms all the tissues of the developing embryo. During
development of the embryo, germinal stem cells are formed,
which persist in adult to allow the cycle of life to continue. In


                                 1
the adult, tissue is renewed by proliferation of specialized stem
cells, which divide to form one cell that remains a stem cell and
another cell that begins the process of differentiation to
specialized function of a mature cell type, normal tissue renewal
is accomplished by the differentiating progeny of stem cells, the
so-called transit amplifying cells. For example, blood cells are
mature cells derived from hematopoietic stem cells in the bone
marrow; the lining cells of the gastrointestinal tract are formed
from transit amplifying cells, progeny of stem cell in the base of
intestinal glands (Crosta, 2010).
   Nineteenth century pathologists first hypothesized the
presence of stem cells in the adult as “embryonal rests” to
explain the cellular origin of cancer and the studies indicate that
the most cancers arise from stem cells or their immediate
progeny, the transit-amplifying cells. Cancer results from an
imbalance between the rate at which cells are produced and the
rate at which they terminally differentiate or die. Understanding
how to control the proliferation and differentiation of stem cells
and their progeny is not only the key to controlling and treating
cancer, but also to cell replacement and gene therapy for many
metabolic, degenerative, and immunological diseases (Virchow,
1985).

Stem cell properties:

   Stem cells have a capacity for self-renewal giving rise to
more stem cells, and the ability to differentiate into tissues of

                                2
various lineages under appropriate conditions. They may be
totipotent, pluripotent or multipotent, depending on type. Only
the embryo is totipotent. Embryonic stem cells (ESCs) are
pluripotent, as they are capable of differentiating into many
tissue types, whereas differentiation of adult stem cells is
generally restricted to the tissue in which they reside, as with
hepatocytes in the liver, and haemopoietic stem cells in blood
(figure 1) (Bongso and Lee, 2005).




       Figure (1): Stem cell self-renewal and differentiation (Bongso and
Lee, 2005).




                                  3
A) Stem cell self renewal:
     The defining feature of a true stem cell is the capacity for
self-renewal. Self renewal occurs when a cell that has been
activated to divide does so asymmetrically. The result produces
one cell that is exactly like the mother cell and one cell that
takes on biological functions that are different from those of the
mother cell. Without self-renewal, each activation event would
result in the progressive loss of the originating stem cell
population (Andeson et al., 2001).

B) The stem cell life cycle:

     Stem cell activation is generally followed by a clonal
expansion of the daughter cell that is produced. This is
associated with a series of biological processes that include
proliferation, migration, differentiation, and at some point cell
death. Regulation of these downstream events determines the
net effect that, each stem cell activation has on new tissue
formation (Song et al., 2007).

     C) Stem cell plasticity:
     The term plasticity means that a stem cell from one adult
tissue can generate the differentiated cell types of another tissue.
At this time, there is no formally accepted name for this
phenomenon in the scientific literature. It is variously referred
to     as    “plastisity”   “unorthodox       differentition”    or
“transdifferentiation” (figure 2) (Joanna et al., 2009).



                                 4
To show that the adult stem cells can generate other cell
types requires them to be tracked in their new environment,
whether it is in vitro or in vivo. In general, this has been
accomplished by obtaining the stem cells from a mouse that has
been genetically engineered to express a molecular tag in all its
cells. It is then necessary to show that the labeled adult stem
cells    have    adopted     key       structural   and    biochemical
characteristics of the new tissue they are claimed to have
generated (Gussoni et al., 2002).
   Also it is necessary to demonstrate that the cells can
integrate into their new tissue environment, survive in the tissue,
and function like the mature cells may assume the characteristic
of cells that have developed from the same primary germ layer
or a different germ layer, for example, much plasticity
experiments involve stem cells derived from bone marrow,
which is a mesodermal derivative. The bone marrow stem cells
may then differentiate into another mesodermally derived tissue
such as skeletal muscle, cardiac muscle or liver (Kocher et al.,
2001).
   Stem cell lineage differentiation and commitment is
conventionally               viewed        as   progressively
           downstream, unidirectional and irreversible. The
notion of unidirectional tissue-lineage commitment of stem cells
is being challenged by evidence of plasticity, or lineage
conversion, in adult stem cells. Mechanisms           allowing for
           such plasticity     include trans-             differentiation
which describes the conversion of a cell of one
                                   5
tissue lineage into a cell of an entirely distinct lineage, with
concomitant loss of the tissue-specific markers and function of
the original cell type, and acquisition of markers and function of
the trans-differentiated cell type (Bianco et al., 2005).
   Alternatively, adult stem cell may differentiate into a tissue
that, during normal embryonic development, would arise from a
different germ layer. For example, bone marrow derived cells
may differentiate into neural tissue, which is derived from
embryonic ectoderm and neural stem cell lines cultured from
adult brain tissue may differentiate to form hematopoietic cells,




   Figure (2): Evidence of plasticity of stem cell (Joanna et al., 2009).




                                    6
or even give rise to many different cell types in embryo. In
both cases cited above, the cells would be deemed to show
plasticity, but in the case of bone-marrow stem cells generating
brain cells, the finding is less predictable (Song et al., 2007).
   Alternative mechanisms for explaining apparent stem cell
plasticity involve cell-cell fusion between a stem cell and a
tissue specific cell, the existence of multiple stem cell
populations in one pool of cells, and the ability of the stem cells
to differentiate to a more primitive, less specialized cell lineage,
and then re-differentiate down another lineage (Bongso and
Lee, 2005).
The differentiation potential of stem cells:
   Many of the terms used to define stem cells depend on the
behavior of the cells in the intact organism (in vivo), under
specific laboratory conditions (in vitro), or after transplantation
in vivo, often to a tissue that is different from the one from
which the stem cells were derived (Joanna et al., 2009).
   So they are three classes of stem cells exist: totipotent,
pluripotent multipotent and unipotent.
1) Totipotent:
   Totipotency is the ability of a cell to divide and produce all
of the undifferentiated cells within an organism, from the Latin
word totus, meaning entire; For example, the fertilized egg is
said to be totipotent, because it has the potential to generate all
the cells and tissues that make up an embryo and that support its
development in uterus. After fertilization, the cell begins to


                                 7
divide and produce other totipotent cells; these totipotent cells
begin to specialize within a few days after fertilization. The
totipotent cells specialize into pluripotent cells, which they
develop into the tissues of the developing body. Pluripotent
cells can further divide and specialize into multipotent cells,
which produce cells of a particular function (Svendsen and
Ebert, 2008).
   Adult mammals, including humans, consist of more than 200
kinds of cells. These include nerve cells (neurons), muscle cells
(myocytes), skin (epithelial) cells, blood cells (erythrocytes,
monocytes, lymphocytes, etc.), bone cells (osteocytes) and
cartilage cells (chondrocytes). Other cells, which are essential
for embryonic development but are not incorporated into the
body of the embryo, include the extraembryonic tissues,
placenta, and umbilical cord. All of these cells are generated
from a single, totipotent cell, the zygote or fertilized egg
(Joanna et al., 2009).


2) Pluripotent:
   Pluripotent stem cells can give rise to any type of cell in the
body except those needed to develop a fetus or adult because
they lack the potential to support the extraembryonic tissue
(e.g., the placenta). Most scientist use the term pluripotent to
describe stem cells that can give rise to cells derived from all
three embryonic germ layers (endoderm, mesoderm, and




                               8
ectoderm). These three germ layers are the embryonic source of
all cells of the body (figure 3) (Svendsen and Ebert, 2008).




   Figure (3): Pluripotent stem cells (Svendsen and Ebert, 2008).



   The term “pluri” is derived from the Latin word plures,
means several or many. Thus, pluripotent cells have the
potential to give rise to any type of cell, a property observed in
the natural course of embryonic development and under certain
laboratory conditions. Pluripotent stem cells are isolated from
embryos that are only several days old; cells from these stem




                                   9
cell lines can be cultured in the lab and grown without limit
(Sonja et al., 2006).
3) Multipotent:
   Multipotent cells, in contrast, can only give rise to a small
number of cell types and they can produce only cells of a
closely related family cell. As haematopiotic stem cells that
differentiate to red blood cells, white blood cells and platelets.
A hematopoietic cell, or a blood stem cell, can develop into
several types of blood cells but cannot develop into liver cells or
other types of cells; the differentiation of the cell is limited in
scope. A multipotent blood cell can produce red and white
blood cells (figure 4) (Svendsen and Ebert, 2008).




   Figure (4): Multipotent stem cell (Svendsen and Ebert, 2008).



                                10
4) Unipotent:
   Unipotent stem cells, a term that is usually applied to a cell
in adult organisms, means that the cells in question are capable
of differentiating along only one lineage. The term “uni” is
derived from the Latin word unus, which means one. Also, it
may be that the adult stem cells in many differentiated,
undamaged tissues are typically unipotent and give rise to just
one cell type under normal conditions. This process would
allow for a steady state of self renewal for the tissue. However,
if the tissue becomes damaged and the replacement of multiple
cell types is required, pluripotent stem cells may become
activated to repair the damage (Avasthe et al., 2008).




   F igu re (5): Differentiation of human stem cells (Bongso and Lee,
2005).



                                11
Classification of stem cells according to their sources:
   Stem cells can be classified into four broad types based on
their origin, stem cells from embryos; stem cells from the fetus;
stem cells from umbilical cord; and stem cells from the adult.
Each of these can be grouped into subtypes (Andeson et al.,
2001).


1) Embryonic stem cells:
   In mammals; the fertilized oocyte, zygote, 2-cells, 4-cells, 8-
cells and morula resulting from cleavage of the early embryo
are examples of totipotent cells (ability to form a complete
organism) (figure 6) (Avasthe et al., 2008).




                               12
Figure (6): Development and differentiation of human tissues (Avasthe
   et al., 2008).



   The inner cell mass (ICM) of the 5 to 6 days old human
blastocyte is the source of pluripotent embryonic stem cells
(HESCs) and consisting of 50–150 cells (figure 7) (Bongso and
Lee, 2005).




                                  13
Figure    (7):     Human blastocyst
showing     inner     cell    mass   and
trophectoderm       (Bongso   and    Lee,
2005).




                                      14
Figure (8): How human embryonic stem cells are derived? (Bongso
and Lee, 2005).



Characteristics of human embryonic stem cells:
   They can maintain undifferentiated phenotype and these
cells are able to renew themselves continuously through many
passages leading to the claim that they are immortal, also these



                                15
cells are pluripotent, meaning that they are able to create all
three germ layers of the developing embryo and thus they can
develop into each of the more than 200 cell types of the adult
body (figure 9) (Junying et al., 2006).




   Figure (9): Characteristics of embryonic stem cells (Junying et al.,
2006).


                                 16
Nearly all research to date has taken place using mouse
embryonic stem cells (MES) or Human embryonic stem cells
(HESCs). Both have the essential stem cell characteristics, yet
they require very different environments in order to maintain an
undifferentiated state. Mouse ES cells are grown on a layer of
gelatin and require the presence of Leukemia Inhibitory Factor
(LIF) (Bongso and Lee, 2005).
    HESCs are grown on a feeder layer of mouse embryonic
fibroblasts (MEFs) and require the presence of basic Fibroblast
Growth Factor (bFGF or FGF-2). Without optimal culture
conditions or genetic manipulation, embryonic stem cells will
rapidly differentiate (Avasthe et al., 2008).


Identification of the human embryonic stem cells:
    Laboratories that grow human embryonic stem cell lines use
several kinds of tests to identify the human embryonic stem
cells.
These tests include:
    1- Growing and sub-culturing the stem cells for many
         months. This ensures that the cells are capable of long
         term self-renewal. Scientists inspect the cultures through
         a microscope to see that the cells look healthy and
         remain undifferentiated (Lawrence et al., 2006).
    2- Using specific techniques to determine the presence of
         surface markers that are found only on undifferentiated
         cells. Another important test is for the presence of a


                                 17
protein called oct-4,      which    undifferentiated cells
       typically make. Oct-4 is a transcription factor, meaning
       that it helps turn genes on and off at the right time, which
       is an important part of          the processes       of   cell
       differentiation and embryonic development.
   3- Examining the chromosomes under a microscope. This is
       a method to assess whether the chromosomes are
       damaged or if the number of chromosomes has changed.
       It does not detect genetic mutations in the cells.
   4- Determining whether the cells can be subculture after
       freezing, thawing, replanting (junying et al., 2006).
Differentiation of human embryonic stem cells:
   In order to start differentiation, the HESCs must be removed
from the feeder layer and the cell replated and will form
embryoid bodies (Ebs), spherical aggregates in which the
HESCs undergo mixed spontaneous differentiation toward
lineages of all three dermal layers. Another protocol of
differentiation directly without formation of embryoid bodies
stage have resulted in more controlled differentiation and better
yield of the required cells (figure 10) (Joanna et al., 2009).




                                18
Figure (10): Fluorescent markers can be used to identify stem cells hidden
among ordinary adult cells. Here, human embryonic stem cells are
recognized by the marker proteins they express (green) (Joanna et al.,
2009).


Ethical considerations:
   The promise of stem cell therapy has ignited public dispute
on the ethics of using aborted embryos for medical purposes.
Individual attitudes are usually influenced by religious and
liberal views but also by concerns that the practice of embryonic
tissue transplantation will increase the pressure to perform
abortions and create a black market in which pregnancy and
aborted tissues will be sold to the highest bidder. The regulated
banking of stem cell lines may solve some of the ethical issues.
As in other cases in which medical and scientific advances
found society without the means to deal with their ethical, legal,
and social consequences, it is important to discuss these issues
in public, with the active participation of the medical and
scientific community (Christopher, 2008).




                                   19
2) Fetal stem cells:
   The identification of human fetal stem cells has raised the
possibility of using autologus cells for in utero treatments. The
human fetal stem cells population extracted from fetal blood
contains adherent cells that divide in culture for 20 to 40
passages and can differentiate into mesenchymal lineages
including bone and cartilage, but also have the ability to form
oligodendrocytes and hematopoiotic cells. These cells, which
can be found circulating only during the first trimester, are
similar to hematopoiotic populations in fetal liver and bone
marrow (Avasthe et al., 2008).

3) Umbilical cord stem cells:
   These are cells harvested from the cord blood. Cord blood is
rich in the stem cells and after appropriate human leukocyte
antigen [HLA] matching may be used to treat a variety of
conditions. Characteristics of these cells are identical to adult
stem cells except that they are not derived from adults and that
their concentration is far more in umbilical blood as compared
to adults. The use of umbilical cord stem cells in orthopedics is
still in a nascent stage and most studies currently focus on the
use of the stem cell (Crosta, 2010).

4) Adult stem cell:
   It is an undifferentiated cell that is found in a differentiated
tissue, it can renew itself and become specialized to yield all the


                                 20
specialized cell types of the tissue from which it originated.
Adult stem cells, like all stem cells, share at least two
characteristics. First, they can make identical copies of
themselves for long period of time; this ability to proliferate is
referred to as long term self renewal. Second, they can give rise
to mature cell types that have characteristic morphologies
(shapes) and specialized functions (Charron et al., 2009).
   Typically, stem cells generate an intermediate cell type or
types before they achieve their fully differentiated state. The
intermediate cell is called a precursor cells in fetal or adult
tissues are partially differentiated cells that divide and give rise
to differentiated cells. Such cells are usually regarded as
“committed” to differentiating along a particular cellular
development pathway, although this characteristic may not be as
definitive as once thought (Bianco et al., 2005).

   Adult stem cells are rare. Their primary functions are to
maintain the steady state functioning of a cell, called
(homeostasis) and with limitation to replace cells that die due to
injury or disease. For example, only an estimated 1 in 10,000 to
15,000 cells in the bone marrow is a hematopoietic (blood-
forming) stem cell (HSC). Furthermore, adult stem cells are
dispersed in tissues throughout the nature of animal and behave
very differently, depending on their local environment. For
example, HSCs are constantly being generated in the bone
marrow where they differentiate into mature types of blood



                                21
cells. Indeed, the primary role of HSCs is to replace blood cells
(Abdallah and Kassem 2008).
   Unlike embryonic stem cells, which are defined by their
origin (the inner cell mass of the blastocyte), adult stem cells
share no such definitive means of characterization. In fact, no
one knows the origin of adult stem cells in any mature tissue.
Some have proposed that stem cells are somehow set aside
during fetal development and restrained from differentiating.
Definition of adult stem cells vary in the scientific literature
range from a simple description of the cells to a rigorous set of
experimental criteria that must be met before characterizing a
particular cell as an adult stem cell. Most of the information
about adult stem cells comes from studies of mice. The list of
adult tissues reported to contain stem cells is growing and
includes bone marrow, peripheral blood, brain, spinal cord,
dental pulp, blood vessels, skeletal muscle, epithelia of skin and
digestive   system,    cornea,   retina,   liver,   and   pancreas
(Christopher, 2008).
   Ideally, adult stem cells should also be clonogenic. In other
words, a single adult stem cell should be able to generate a line
of genetically identical cells, which then gives rise to all the
appropriate, differentiated cell types of the tissue in which it
resides. Again, this property is difficult to demonstrate in vivo;
in practice, scientists show either that a stem cell is clonogenic
in vitro, or that a purified population of candidate stem cells can
repopulate the tissue (Avasthe et al., 2008).


                                 22
Sources of adult stem cells:
(I) Bone Marrow-Derived Stem/Progenitor Cells:
   Adult bone marrow-derived stem cells are presently the cell
types most widely used in stem cell therapy. A heterogeneous
subset there of, termed autologous bone marrow-derived
mononuclear cells (ABMMNCs), comprises the following types
of stem cells, (Mesenchymal stem cells, Hematopoietic stem
cells and Endothelial progenitor cells), that have potential
therapeutic uses (figure 11) (Svendsen and Ebert, 2008).




Figure (11): Some of the known sources of adult stem cells (Svendsen and
                             Ebert, 2008).




                                  23
(a) Mesenchymal stem cells (MSCs):
   MSCs are a proper stem cell which can be greatly and
efficiently expanded in culture and can differentiate to several
specific mesenchymal cell lineages. Mesenchymal (Stromal)
stem cells (MSCs) are found in various niches of adult tissue.
MSCs are rare in bone marrow (<0.01% of nucleated cells, by
some estimates) and 10 times less abundant than hematopoietic
progenitor cells but MSCs can be readily grown in culture.
However, more recently, other sources of MSCs have been
described including placenta, adipose tissue, cord blood and
liver (junying et al., 2006).
   The human Mesenchymal stem cells (HMSCs) from bone
marrow can be cloned and expanded in vitro more than 1
million-fold and retain the ability to differentiate to several
mesenchymal lineages. Researchers have not yet found
conditions that allow continuous, indefinite HMSC growth, yet
it is possible to produce billions of MSCs in vitro for cellular
therapy from a modest bone marrow aspirate drawn through the
skin. MSCs need to be expanded ex vivo because they
apparently are very contact inhibited, and there is little evidence
of in vivo expansion as MSCs labeled with membrane dyes, that
would be diluted and undetected from dividing cells after about
3 divisions, are found months later even in repairing tissue
(Sottile et al., 2002).




                                24
Advantages of Mesenchymal Stem Cells:
   Ease of isolation, high expansion potential, genetic stability,
reproducible characteristics in widely dispersed laboratories,
compatibility with tissue engineering principles and potential to
enhance repair in many vital tissues. There they may be the
current preferred stem cells model for cellular therapeutic
development (Diana and Gabriel, 2008).
Biology of mesenchymal stem cells (MSCs):
   The anatomical locations of phenotype of MSCs have no yet
been well defined in vivo. Some have used expression of Stro-1
and VCAM-1 to analyses putative MSC in vivo in human. A
general consensus among researchers in the field is that MSC
can be successfully defined based on staining with surface
markers such as CD44, CD90, CD73, CD105 and CD166.
However, none of these antigens are unique to MSC. Using
markers such as Stro-1 (human) and Sca-1 (mouse), several
reports indicate that MSC reside adjacent to endothelium in the
bone marrow and possibly other tissues (Zannettino et al.,
2007).
(b) Hematopoietic stem cells (HSCs):
   HSCs are presented in umbilical cord blood with a frequency
of just under one in 1 million mononuclear cells (one in 3
million MNCs) or mobilized peripheral blood (one in 6 million
MNCs). They are capable of unlimited cell proliferation in bone
marrow and must undergo at least 20 to 23 divisions on their




                               25
way to produce mature blood cells, even assuming no cell death
along the way (Emerson et al., 2008).
Biology of heamtopiotic stem cells:
   Much effort has been focused on discovering cell surface
markers that can identify those cells that have true functional
stem cell properties. Perhaps clinically most familiar is CD34, a
glycoprotein present on the cell surface of stem and progenitor
cells which is used to enrich stem cells mobilization and
collection for HSCs, but even within the CD34+ population,
only a small percentage are HSCs (Emerson et al., 2008).
   For decade scientists and hematologists have struggled with
the difficulty that HSCs cannot be purified based on
phenotypical characteristics and perhaps more importantly,
cannot be expanded and cloned ex vivo. Recent evidence has
emerged suggesting that HSCs can be expanded ex vivo. But
there is still no evidence to support the idea of clonality. For
these reasons HSCs are not ideally suited for in vitro
experiments designed to test plasticity. In this regard HSCs
differ dramatically from MSCs in bone marrow and neural stem
cells (NSCs) in the central nervous system, both of which can
be clonally derived and tested for multiple differentiation
pathways (figure 12) (Joanna et., 2009).




                               26
Figure (12): Hematopoietic and stromal stem cell differentiation (Joanna et al., 2009).
     (c) Endothelial progenitor cells (EPCs):
         A subset of bone marrow-derived hematopoietic progenitor
     cells: endothelial progenitor cells (EPCs). These cells can give
     rise to endothelial recovery and new capillary formation after
     ischemia (Einstein and Ben-Hur, 2008).
      (II) Neural stem cells:
         The concept that adult mammalian CNS contains NSCs was
     first inferred from evidence of neuronal turnover in olfactory



                                          27
bulb and hippocampus in the adult organism. The multipotency
of NSCs was demonstrated in vitro in 1990 by their ability to
differentiate into neurons, astrocytes, and oligodendrocytes as
well as various forms of neural precursors. In addition, in vivo
delivery of these cells to animal models of neurodegenerative
diseases was associated with varying degrees of functional
recovery. Currently, there is no set of markers or protein
expression profiles that precisely define and fully characterize
undifferentiated NSCs. Neural stem cells (NSCs) and neural
precursor cells (NPCs) can be isolated from the developing or
adult CNS and can be safely expanded in chemically defined
culture media for an extended (Song et al., 2007).
(a) Adult neural precursor cells (NPCs):
   New neurons are derived in adulthood from a population of
adult NPCs, which are primarily found in the subependymal
layer of the ventricular zone and the dentate gyrus of the
hippocampus, although they are also probably found in other
sites. However, the behavior of the neural precursor cells
(NPCs) found in all these sites is different, and may relate as
much to the environment in which they find themselves as to
their intrinsic properties, eg; nigral NPCs appear to only
differentiate into astrocytes in situ or when grafted to the adult
nigra, but when they are cultured in vitro or transplanted into the
hippocampus they can form neurons (Gronthos et al., 2003).




                                28
Properties of neural stem cells:
(1) Immunosupressive effect of NSCs:
     Although NSCs may exert their therapeutic effects by
directly replacing missing cells, transplantation rarely results in
significant    numbers       of       transplant-derived      terminally
differentiated neurons. The beneficial effect of NSCs in disease
models may be attributable to alternative biologic properties.
The first indication of an anti-inflammatory effect of NPCs
came from transplantation experiments in rats with experimental
autoimmune         encephalomyelitis         (EAE).   It   was      shown
transplantation of NPCs reduced brain inflammation and clinical
disease severity, it was suggested that the benefit of NPC
transplantation was mediated by an anti-inflammatory effect
(Raisman and Li, 2007).
     The exact mechanisms by which transplanted NPCs
attenuate brain inflammation are unclear. Some suggests an
immunomodulatory effect by which NPCs promote apoptosis of
type 1 T-helper cells, shifting the inflammatory process in the
brain toward a more favorable climate of dominant type 2 T-
helper    cells.     Alternatively,      a     nonspecific    bystander
immunosuppressive effect of NPCs on T-cell activation and
proliferation has been suggested. The suppressive effect of
NPCs on T cells was accompanied by a significant suppression
of    pro-inflammatory      cytokines.        This    nonspecific    anti-
inflammatory mechanism may be of major importance in the
application of transplantation therapy in immune-mediated


                                  29
diseases because it can protect the host CNS and graft from
additional immune attacks (Einstein and Ben-Hur, 2008).
(2) Neuroprotictive effects of transplanted NSCs:
   Neuroprotective effect     was    observed    in    other   non
autoimmune experimental disease models. Neural stem cells
rescued dopaminergic neurons of the mesostriatal system in a
Parkinson disease (PD) model in rodents. These findings led to
the concept that NSCs are endowed with inherent mechanisms
for rescuing dysfunctional neurons. This effect was found to be
important in other neurologic diseases. Neural stem cells seeded
on a synthetic biodegradable scaffold and grafted into the hemi-
sectioned adult rat spinal cord induced significant improvement
in animal movement by reduction of necrosis in the surrounding
parenchyma and by prevention of inflammation, glial scar
formation, and extensive secondary cell loss (Einstein and Ben-
Hur, 2008).
(3) Neurotrophic effects of transplanted NSCs:
   After   sectioning   of   the    adult   spinal    cord,    NSC
transplantation induced a permissive environment for axonal
regeneration. Similarly, in a model of retinal degeneration, NPC
transplantation promoted neural growth in the optic nerve. In
both cases, this effect was mediated by induction of matrix
metalloproteinases that degrade the impeding extracellular
matrix and cell surface molecules, enabling axons to extend
through the glial scar. Transplantation of olfactory-ensheathing
cells into the sectioned spinal cord also promoted axonal


                               30
regeneration in long fiber tracts, with a return of lost function.
This was explained by the creation of proper realignment,
enabling axonal growth through a permissive tract. In addition,
the cells increased axonal sprouting, remyelination, and
vascularization of the injured spinal cord (Raisman and Li,
2007).
Isolation of human NSCs:
   To date, they are primary isolated and propagated in vitro as
cells that form free-floating neurospheres when cultured in
serum-free medium on non adherent surfaces in the presence of
mitogenic factors such as basic FGF or FGF-2 and epidermal
growth factors, although there have also been reports of
monolayer cultures (McBride et al., 2004).
(III) Pancreatic stem/progenitor cells:
   There is strong evidence that new pancreatic islets can
derive from progenitor cells present within the ducts and islets,
in a process called “neogenesis”. Furthermore, when these
pseudo-islets were transplanted into non-obese diabetic (NOD)
mice, diabetes reversal was observed. Candidate pancreatic
stem/progenitor cells have also been described within acini, but
contamination with endocrine and ductal cells in cultures could
not be excluded in these experiments (Limmbert et al., 2008).
   The isolation of a distinct stem/progenitor cell within the
endocrine pancreas depends on the identification of a specific
progenitor marker. The exciting observation that nestin positive
islets cells display endocrine differentiating capacity led to the


                                31
hypothesis that this intracytoplasmic filament protein might
correspond to a pancreatic stem/progenitor cell marker. More
recently, in two important studies a population of cells in the
developing and adult mouse pancreas was identified, which
under differentiation conditions, released insulin in a glucose-
dependant manner. After differentiation, these cells expressed
specific developmental pancreatic endocrine genes (e.g. Ngn3,
Pax-4, Pax-6 and PDX-1) and contamination with mature beta
cells was ruled out (Limbert et al., 2008).
   While mature beta cell replication appears to be major
physiological beta-cell regenerative process, identification of
pancreatic cells with progenitor features might open an
important and promising strategy for cell replacement and
regeneration therapy. Anyhow, to be clinically relevant, in vitro
proliferation of progenitor cells from human pancreas must
produce large amounts of cells, in order to allow cells isolated
from one single donor to be sufficient to treat a given diabetic
patient. It would be even better to have one single donor for
several   diabetics.    For   these    reasons,     acinar     isolated
stem/progenitor cells might be of interest, considering that
exocrine tissue constitutes 90% of pancreatic tissue and is
discarded during islet isolation (Kushner et al., 2005).
(IV) Other sites:
   First identified in human bone marrow, a population of
mesenchymal      progenitor/stem      cells     (MSC)    with     well
characterized       immunophenotype           and   distinct     from


                                32
hematopoietic stem cells, was shown to possess a high
        proliferation rate and great plasticity. Under specific culture
        conditions these cells differentiate into mesenchymal tissues,
        such as bone, cartilage, muscle, tendon, adipose and stroma, as
        well as neuronectodermal tissues (Limbert et al., 2008).
           Adult tissues and organs known to have stem cells
Source            Description
Brian             Stem cells of the brain can differentiate into the three kinds of
                  nervous tissue-astrocytes, oligodendrocytes, and neurons-and in
                  some cases, blood-cell precursor.

Bone marrow       These occur as hematopoietic stem cells, which give rise to all blood
                  cells, and as stroma cells, which differentiate into cartilage and bone.

Endothelium       These stem cells are called hemangioblasts and are known to
                  differentiate into blood vessels and cardiomyocytes. They may
                  originate in bone marrow, but this is uncertain.

Skeletal muscle   These stem cells may be isolated from muscle or bone marrow. They
                  mediate muscle growth and may proliferate in response to injury or
                  exercise.

Skin              Stem cells of the skin are associated with the epithelial cells, hair
                  follicle cells, and the basal layer of the epidermis. These stem cells
                  are involved in repair and replacement of all types of skin cells.

Digestive         Located in intestinal crypts, or invaginations. These stem cells are
system            responsible for renewing the epithelial lining of the gut.

                  Many types are believed to exist, but examples have yet to be
Pancreas          isolated. Some neural cells are known to generate pancreatic β cells.

                 The identity of liver stem cells is still unclear. Stem cells from bone
Liver            marrow may repair some liver damage, but most repairs seems to be
                 carried out by the hepatocytes (liver cells) themselves.
      Table (1): Sources of adult stem cells (Limbert et al., 2008).




                                            33
Identification of the adult stem cells:
   The scientists often use one or more of the following three
methods to identify and test adult stem cells:
1- Labeling the cells in a living tissue with molecular markers
   and then determining the specialized cell types they
   generate. Then
2- Removing the cells from living animals, labeling them in
   cell culture, and transplanting them back into another animal
   to determine whether the cells repopulate their tissue of
   origin. Then
3- Isolating the cells, growing them in cell culture, and
   manipulating them, often by adding growth factors or
   introducing new genes, to determine what differentiated
   cells types they can become (Raisman and Li, 2007).
The similarities and differences between embryonic and adult
stem cells:
   The adult and embryonic stem cells differ in the number and
types of differentiated cells types they can become. Embryonic
stem cells can become all cell types of the body because are
pluripotent. Adult stem cells are          generally limited to
differentiating into different cell types of their tissue of origin.
However, some evidence suggests that adult stem cell plasticity
may exist; increasing the number of cell types a given adult
stem cell can become (figure 13). Large numbers of embryonic
stem cells can be relatively easily grown in culture, while adult
stem cells are rare in mature tissues and methods for expanding


                                34
their numbers in cell culture have not yet been worked out. This
is an important distinction, as a large number of cells are needed
for stem cell replacement therapies (Limbert et al., 2008).
    A potential advantage of using stem cells from an adult is
that the patient’s own cells could be expanded in culture and
then reintroduced into the patient. The use of patient’s own
adult stem cells would means that the cell would not be rejected
by the immune system. This represents a significant advantage
as immune rejection is a difficult problem that can only be
circumvented with immunosuppressive drugs. Embryonic stem
cells from a donor introduced into a patient could cause
transplant rejection, however, whether the recipient would reject
donor embryonic stem cells has not been determined in human
experiments (Sonja et al., 2006).




Figure (13): Sources of stem cells (Limbert et al., 2008).


                                     35
Types of Stem Cell transplantation:
   Stem cell transplantation can be classified according to the
genetic relation between the donor and recipient into 4 classes:
   1- Autograft: In which the donor and recipient is the same
       individual.
   2- Isograft or syngenic graft: In which the donor and
       recipient are genetically identical (e.g., monozygotic
       twins).
   3- Allograft or homograft: In which the donor and recipient
       are genetically unrelated but belong to the same species.
   4- Xenograft or heterograft: In which the donor and
       recipient belong to different species (David, 2009).
Application of stem cells:
   1) Basic science application:
Stem cells are ideally suited to allow for the study of complex
processes that direct early unspecialized cells to differentiate
and develop into the more than two hundred cell types in the
human body (Bianco et al., 2005).
   2) Medical research applications:
Stem cell studies may allow researchers to follow the processes
by which diseases and impairments caused by genetic
abnormalities first    manifest     themselves   biochemical   or
structurally in cells and tissues. Using stem cells to produce
large numbers of genetically uniform cultures of organ tissues
for example, liver, muscle, or neural would allow controlled
comparison of the effects of drugs or chemical on these tissues.


                               36
Alternatively, testing drugs against stem cell tissues varying
genetic makeup could allow tissue specific stem cell may
provide a constant in vitro source of such cellular material
(Bianco et al., 2005).
The site of stem cell implantation:
   The transplantation can be described as orthotropic or
heterotropic:
   1- Neurologic transplantation: Refers to donor tissue
       implantation in the anatomically correct position in the
       recipient.
   2- Heterotropic transplantation: Refers to the relocation of
       the implant in the recipient at a site different from the
       normal anatomy (David, 2009).
Route of stem cell delivery:
   Reports      have     indicated    that   after   stereotactic
intraparenchymal, intracerebro-ventricular, intravenous      and
intraarterial transplantation, stem cells can home to sites of
injury in the CNS and induce functional recovery. Of these
various transplantation techniques, those that depend on
intravascular delivery of stem cells for stroke are particularly
attractive.
Intravascular delivery:
   In addition to its minimal invasive nature, intravascular
delivery may allow stem cells to have a superior interaction
with injured tissue. A comparative study revealed that direct
intracerebral transplantations resulted in the largest number of


                                37
cells at the lesion site, followed by intracerebro-ventricular and
intravenous transplantations (Guzman et al., 2008).
   However, researchers in that study only assessed the
absolute number of cells in the perilesional area and took no
account of whether these cells were therapeutically distributing
to all injured areas of brain parenchyma on a microscopic level.
Many believe that intravascular delivery of stem cells may lead
to a wider distribution of cells around the lesion as compared
with focal perilesional transplants, thereby leading to superior
stem cell–injured tissue interactions (Xiao et al., 2007).
Mechanism of wide distribution:
   The cells travel in the blood stream and follow a chemo
attractant gradient generated by inflammation in the injured
brain. Unfortunately, intravenously delivered cells pass through
the systemic and pulmonary circulation systems and home to
other organs as well, which significantly reduces cell homing to
the injured brain. Intravenous injection of human MSCs into
rats 24 hours after stroke showed that only 4% of the cells
entered the brain, the number of cells entering the brain
increased over time and peaked at Day 21 post-stroke. At Day
56, 60% of these surviving cells differentiated into glia, and
20% into neurons. Despite the fact that the number of cells
entering the brain was limited, functional recovery was
enhanced by intravenous delivery (Pluchino et al., 2005).




                                38
Intracarotid injection:
   Another route of intravascular delivery is intra arterial,
which would circumvent body circulation. The first pass of stem
cells injected into the carotid artery would be the brain, this
route of delivery have demonstrated functional recovery after
stroke and traumatic brain injury. In 2006 Shen and colleagues
injected donor rat BMSCs into the internal carotid artery of rats
24 hours post-stroke and successfully induced functional
recovery. In another study, the same group injected donor rat
BMSCs into rats 24 hours after stroke and observed that
injected cells localized around the infarction area in the brain
and very few were found in the heart, lungs, liver, spleen, and
kidney (figure 14) ( Guzman et al., 2008).




Figure (14): Confocal laser scanning microscopy images revealing
numerous cells in the stroke border zone and the hippocampus ipsilateral to
the stroke (A). Inset shows doublecortin bromodeoxyuridine labeled cells.
The VCAM-1 (arrows) is highly expressed in the stroke affected
hemisphere 48 hours after stroke (B). DCX = doublecortin; BrdU =
bromodeoxyuridine; DAPI = 4'6-diamidino-2-phenylindole (Guzman et al.,
2008).


                                   39
The debate over the best delivery route is further
complicated by the fact that there is still a great deal of
controversy concerning the mechanism by which stem cells lead
to enhanced functional recovery in patients who have
experienced stroke. The 2 most discussed mechanisms are as
follows: 1) cellular replacement, by way of the functional
integration of stem cells; and 2) secretion of neurotrophic and
angiogenic factors. If the mechanism of recovery is cellular
replacement, then transendothelial migration is necessary and
the methods that allow the highest concentrations of stem cells
in the injured brain areas ought to be pursued; however, there is
significant evidence that stem cells may provide their benefits
by secreting various neuroprotective factors (Guzman et al.,
2008).
   In summary, the best route of human stem cell delivery has
not been determined, but the intravascular route is particularly
attractive because of its ease of administration, minimal
invasiveness, and potential for widespread cell distribution
together   with   widespread    secretion   of   neuroprotective,
proangiogenic, and immunomodulatory factors. Intuitively, the
intraarterial route of delivery seems better than the intravenous,
given that injected cells first pass the target organ that is, the
brain prior to being redistributed in the systemic circulation
(Pluchino et al., 2005).




                               40
Timing of transplantation
   Undoubtedly the fate and function of transplanted cells will
depend on any or all of these alterations, and the optimal time of
transplantation is unknown. The timing of transplantation
depends mainly on the goal of treatment, for example,
neuroprotection, which should happen early after the insult, or
neuroregeneration/cell replacement, which can be done once a
lesion has stabilized. We can envision a future in which we will
rely on multimodal stem cell treatment, depending on a
combination of early and late administrations of different cell
types (Guzman et al., 2008).
Early intravascular cell delivery
   In animal models with a neuro inflammatory component
such as stroke, traumatic brain injury, spinal cord injury, and
multiple sclerosis, therapeutic somatic stem cells (for example,
BMSCs, umbilical cord blood stem cells, MSCs, and NPCs)
target inflamed CNS areas where they persist for months and
promote recovery through neuroprotective mechanisms. It is
thought that the process of transendothelial migration of somatic
stem cells may be regulated in a manner similar to that of
inflammatory cells. As early as 30 minutes after stroke, the
infiltration of leukocytes, both polymorphonuclear leukocytes
and monocytes/macrophages, can be observed (Goldman and
Windrem, 2006).
   Chemoattraction, adhesion, and transendothelial migration
of inflammatory cells is regulated by specific inflammatory


                               41
mediators, which have been identified in experimental and
human stroke. The temporal expression profile of adhesion
molecules, cytokines, and chemokines after stroke has been well
described. Vascular cell adhesion molecule–1 (VCAM-1) has
been shown to reach a peak level 24 hours after experimental
stroke. At the bedside, soluble VCAM-1 concentration in
plasma is increased in patients with acute stroke. Intercellular
adhesion molecule–1 levels have been elevated as early as 4
hours after stroke with sustained levels for up to 1 week (Zhang
and Lodish, 2005).
   Monocyte chemo-attractant protein–1, a key chemoattractant
factor for the recruitment of circulating peripheral cells to the
stroke area and an important factor for stem cell migration, is
upregulated 3 days after stroke and then returns to baseline after
1 week. Similarly, stromal-derived factor–1(SDF-1) is known to
be a potent chemoattractant for inflammatory as well as stem
cells (including BMSCs and NSCs) and is expressed early after
stroke. Anatomically, adhesion molecule upregulation as well as
chemokine expression has been shown to be highest in the
stroke-affected penumbral region. Blocking the different
pathways of chemoattraction and cell adhesion in stroke-
affected   rodents   reduced     the   number     of   infiltrating
inflammatory cells (Belmadani et al., 2006).
   In mice lacking intercellular adhesion molecule–1 (ICAM-
1), a significant reduction in inflammatory cellular infiltrate and
a reduction in lesion size were noted. Treatment with anti–


                                42
ICAM-1 antibodies was a successful neuroprotective means of
reducing lesion size and apoptosis in experimental stroke.
However, a clinical trial exploring the feasibility of using an
ICAM-1 blocking antibody failed to demonstrate any beneficial
effects in the patients. There is some evidence                  that
intravascularly administered stem cells undergo the same
process as inflammatory cells, including chemoattraction,
adhesion,     and    transendo-thelial    migration   after   stroke,
potentially making this route an ideal way of cell delivery (Hill
et al., 2004).
Late intraparenchymal cell transplantation
   In contrast to the acute intravascular cell treatment, the
intraparenchymal approach has been hindered by poor outcomes
if the stem cells are transplanted too early after stroke.
Excitotoxicity, oxidative stress, and inflammation post ischemia
make    the      ischemic   brain    a   hostile   environment    for
intracerebrally transplanted cells. In fact, we have found a
negative correlation between graft survival and inflammation.
Additionally, we demonstrated that human NSCs transplanted
too close or into the stroke area have very limited survival at
days after stroke (Belmadani et al., 2006).
   Transplanting cells 3 weeks after stroke, when there is a
significant decrease in inflammation, led to greater graft
survival than transplanting 5–7 days after stroke. Taken
together, early intravascular cell therapy might benefit from the
processes tied to post-stroke inflammation but might be


                                    43
detrimental to cells directly transplanted intra-parenchymally.
Therefore, intra-parenchymal cell replacement therapy might be
useful as a second line or delayed stem cell treatment strategy
(Grabowski, 2010).




                              44
Stem cell therapy in Parkinsonism

   Parkinson’s disease (PD) otherwise known as ‘’paralysis
agitans’’ or ‘’shaking palsy’’ was classically described by James
Parkinson in 1817. His description of “Involuntary tremulous
motion with lessened muscular power, in parts not in action and
even when supported, with a propensity to bend the trunk
forward and to pass from a walking to a running pace, the
senses and intellect being uninjured” has stood the test of time.
PD is also defined as a debilitating neurodegenerative disorder
of insidious onset in middle or late age characterized by the
selective loss of nigrostriatal dopaminergic neurons and loss of
dopamine in the striatum (Abayomi, 2002).
   Parkinson’s disease is second only to Alzheimer’s disease
with a prevalence of 1 in 10,000. Although it is uncommon in
people under age 40 years, the incidence of PD greatly increases
with age, affecting approximately 1% of individuals older than
60 years (Lane et al., 2008).
Pathology:
   The basic pathology is cell degeneration and loss of
pigmented neurons in the pars compacta of the substantia nigra
and locus ceruleus with atrophy and glial scarring. The
degenerated pigmented neurons contain Lewy bodies which are
intracytoplasmic eosinophilic hyaline inclusions composed of
protein filaments (ubiquitin & synuclein), and do not have the
electronic microscropic appearance of any known viral or


                                45
infective agent (fig. 15). Lewy bodies are characteristic of
Parkinson’s disease except in post-encephalitic Parkinson’s and
parkengene mutants. However, they could be found in 4% of
brain without parkinsonian features and these are likely cases of
subclinical Parkinson’s as 80% of the zona compacta cells must
degenerate   before    clinical    symptoms   become     apparent
(Abayomi, 2002).
   The pars compacta contains 450,000 dopaminergic neurons.
With the loss of dopaminergic neurons at those sites, there is
deficiency of dopamine in the basal ganglia, chiefly the striatum
(caudate nucleus and putamen). Furthermore, the enzymes
required for dopamine synthesis, DOPA decarboxylase and the
rate limiting enzyme tyrosine hydroxylase are reduced. In
addition, there is deficiency of neurotropic factors such as glial
and brain derived neurotrophic factors. However, neurons in the
striatum with dopamine receptors remain intact and are
responsible for the therapeutic effects of levodopa. In the
Parkinsonism unresponsive to levodopa, striatal neurons are
degenerated. Genetic and environmental factors are important in
the mechanism of neuronal deaths due to neuronal necrosis or
apoptosis. In neuronal necrosis there is disintegration of cell and
organelles and subsequent         removal by phagocytic       and
inflammatory response with increased cellular permeability. In
apoptosis on the other hand, there is rapid programmed cell
death in response to a toxic stimuli. There is chromatin
condensation, DNA fragmentation and cell shrinkage, with


                                  46
relative sparing of organelles without inflammatory changes or
increased cellular permeability (Golbe, 2003).
   Among the factors that have been implicated in neuronal
degeneration    in   Parkinson’s     disease   are   mitochondrial
dysfunction, oxidative stress, and the actions of excitotoxins,
deficient neurotrophic support and immune mechanisms. HLA-
DR positive reactive microglial cells and cytokines such as
interleukin 1 (IL-1) and tumor necrosis factor-a play significant
role in the pathogenesis of Parkinson’s disease. Oxidative stress
with excess reactive oxygen species and free radical damage
involving one or more unpaired electrons react with nucleic
acids, proteins and lipids, this metabolic derangement results in
generation of toxic byproducts and increased oxidative stress
with resultant cellular damage (figure 15) (Lane et al., 2008).




                                47
Figure (15): Neuronal Pathways that degenerate in Parkinson's disease.
Signals that control body movements travel along neurons that project from
the substantia nigra to the caudate nucleus and putamen (collectively called
the striatum) (Lane et al., 2008).


Cell replacement therapy:
   The idea of growing dopamine cells in the laboratory to treat
Parkinson’s is the most recent step in the long history of cell or
tissue transplantation to reverse this devastating disease. The
concept was, and still is, straightforward: implant cells into the
brain that can replace the lost dopamine releasing neurons.
Although conceptually straight forward, this is not an easy task.
Fully developed and differentiated dopamine neurons do not
survive transplantation, so direct transplantation of fully
developed brain tissue from cadavers, for example, is not an


                                    48
option. Moreover, full functional recovery depends on more
than cell survival and dopamine release; transplanted cells must
also make appropriate connections with their normal target
neurons in the striatum (Lindvall and Kokaia, 2010).
   Under the basic principle of restoring dopamine producing
neurons via neural grafts, extensive studies have been done to
bring this to fruition. One of the first attempts at using cell
transplantation in humans was tried in the 1980s. This surgical
approach involved the transplantation of dopamine producing
cells found in the adrenal glands, which sit atop the kidneys in
the abdomen dramatic improvement in Parkinson’s patients by
transplanting dopamine producing chromaffin cells from several
patients’ own adrenal glands to the nigrostriatal area of their
brains; it showed dramatic improvement in Parkinson’s patients.
Another strategy was previously attempted in the 1970s, in
which cells derived from fetal tissue from the mouse substantia
nigra was transplanted into the adult rat eye and found to
develop into mature dopamine neurons (Panchision, 2006).
   The functional integration of dopamine neuron grafts prove
the efficacy of the cell replacement principle, but in reality, this
clinical outcome is extremely inconsistent with respect to the
percentage of cells that survive the grafting procedure and the
amount of dopamine produced by the new neurons. In fact,
average functional improvement of patients in the experiments
only rises about 20%. Across the board, subjects achieve
functioning levels less than or equal to that of patients


                                49
undergoing deep brain stimulation, which carries a lower
morbidity risk (Lane et al., 2008).
Cell transplantation:
   Transplantation of primary ventral mesencephalic tissue into
the striatum aims to restore brain circuitry and function lost as a
result of PD.      The main objective        of primary      tissue
transplantation has been to provide proof of principle that
grafted dopaminergic neurons can i) survive and restore
regulated dopamine release, ii) integrate with the host brain to
reinstate frontal cortical connections and activation, and iii) lead
to measurable clinical benefits together with improved quality
of life. Preclinical work in animal models of PD has shown that
grafted dopaminergic neurons, extracted from the developing
ventral mesencephalon (VM) can survive, reinnervate the
lesioned striatum, and improve motor function (Winkler et al
2005).
   Over the past two decades, a series of open label clinical
trials have provided convincing evidence to show that human
embryonic nigral neurons taken at a stage of development when
they are committed to a dopaminergic phenotype can survive,
integrate and function over a long time in the human brain.
There is good evidence of graft survival, with grafted neurons
developing afferent and efferent projections with the host
neurons. Long term survival of dopaminergic grafts is possible
up to 10 years after transplantation, and there have been no




                                50
reported cases of overt immunorejection even after several years
of withdrawal from immuno-suppression (Olanow et al 2003).
    Evidence from Positron Emission Tomography (PET)
scanning has revealed significant increases in activation in the
areas reinnervated by the grafted cells, and longitudinal clinical
assessments indicate significant functional recovery for motor
control, in some cases for more than 10 years, in the most
successful cases, patients have either reduced dependency for or
completely withdrawn from L-dopa treatment. Post mortem
studies similarly show good survival of transplanted neurons
and well integrated grafts (figure 16) (Winkler et al., 2005).




Figure (16): Dopamine Neuron Transplantation: PET images from a
Parkinson’s patient before and after fetal tissue transplantation, the image
taken before surgery (left) shows uptake of a radioactive form of dopamine
(red) only in the caudate nucleus, indicating that dopamine neurons have
degenerated. Twelve months after surgery, an image from the same patient
(right) reveals increased dopamine function, especially in the putamen
(Winkler et al., 2005).




                                    51
The precise mechanism responsible for these dyskinesia
remains unknown but it does not appear to be related to graft
overgrowth resulting in excessive dopamine release. One
possibility surrounds the quality of dissected tissue. Successful
trials have used either freshly dissected tissue or tissue that has
been stored in culture for only a few days. One of the trials
reporting cases of severe dyskinesias used tissue stored in
culture for up to four weeks and it may be that holding tissue in
this way reduces its dopaminergic composition (Olanow et al.,
2003).
   A further issue concerns the identification of dense
hyperdopaminergic areas within the graft of some patients with
graft induced dyskinesias. This may have caused uneven striatal
innervation   and    excessive    dopamine     release into     non
reinnervated areas. It is also possible that variable side effects of
graft induced dyskinesias are related to patient selection.
Greater functional improvement is associated with younger
patients, and in patients with less advanced disease. This is most
likely because the neuropathology is relatively confined to the
nigrostriatal pathway and may have better trophic support
compared to patients with more advanced disease (Winkler et
al., 2005).
Stem cells:
   Stem cells could provide one such source and would
overcome the issue of limited availability of fresh primary fetal
cells. A wide range of stem cells are being investigated as


                                 52
potential sources of dopaminergic neurons for transplantation,
stem cells can be obtained from various sources (Morizane et
al., 2009).
   The majority of research thus far with respect to the
formation of dopaminergic neurons for the treatment of PD is in
embryonic stem cells and neural stem cells. Dopaminergic
neurons are more easily obtained from neural stem cells in the
developing ventral mesencephalon (VM) than other parts of the
developing central nervous system but the number of
dopaminergic cells produced is still very low. Despite genetic
manipulation and the addition of various growth and
differentiation   factors,   generating    large     numbers    of
dopaminergic cells from this cell type has had mixed results
(Panchision, 2006).
   However, greater success has been achieved with the more
complex ES cells. Derived from blastocysts donated following
in vitro fertilization these cells are truly pluripotent. Promising
data have been obtained with dopaminergic neurons derived
from mouse ES cells, significantly improving motor function in
a rat model of PD. However, directing the differentiation of
human ES cells has proved complex and while 50% of cells
spontaneously differentiate into neurons           upon   leukemia
inhibitory factor (LIF) withdrawal, few are dopaminergic. Thus,
there is the need to develop protocols to ‘direct’ differentiation.
The most successful published protocols describe multiple
culture stages in which different transcription and growth


                                53
factors are added at controlled time points (Goldman and
Windrem, 2006).
   However, despite good yield of dopaminergic neurons in
vitro, clinically relevant long term survival and behavioral
recovery in animal models rivaling that of primary tissue has yet
to be convincingly demonstrated. Neuronal stem cells unlike
embryonic stem cells, which are only derived from the
embryonic blastocyst, neural stem cells can be found both in
embryonic neural tissue and also in specific neurogenic regions
of the adult brain. If the in vivo survival of neural stem cells can
be improved they hold the potential to provide autologous
transplantation as patients provide the cells for their own
recovery (Morizane et al., 2009).
   Interestingly, stem cells may not just be useful as dopamine
factories in the striatum. Some studies in both rodent and
primate models have shown significant behavioral recovery
following transplantation with neural stem cells. In addition to
the generation of a small population of dopaminergic neurons
other cells within the graft were found to be releasing growth
factors which are purportedto            exert neuro-protective or
neuroregenerative influences. While more evidence needs to be
accumulated on the longevity of this effect, it broadens the
potential of    neural stem cells from simple dopamine
replacement    to    preserving        and   enhancing   remaining
dopaminergic neurons (Svendsen and Langston, 2004).




                                  54
Stem cell based approaches could be used to provide
therapeutic benefits in two ways: first, by implanting stem cells
modified to release growth factors, which would protect existing
neurons and/or neurons derived from other stem cell treatments;
and second, by transplanting stem cell derived DA neuron
precursors/neuroblasts into the putamen, where they would
generate new neurons to ameliorate disease-induced motor
impairments (figure 17) (Lindvall and Kokaia, 2010).




Figure (17): Stem cell based therapies for PD. PD leads to the progressive
death of DA neurons in the substantia nigra and decreased DA innervation
of the striatum, primarily the putamen (Lindvall and Kokaia, 2010).


Neurogenesis:
   As mentioned above endogenous stem cells are present in
specific regions of the brain. While the occurrence of
neurogenesis in the striatum and substantia nigra is debated, one


                                   55
indisputable neurogenic region is the subventricular zone (SVZ)
lying adjacent to the striatum. The cells in the region are an
assortment of stem and progenitor cells that have the potential to
be mobilized and induced to differentiate by the presence of
growth factors or other small molecules. In the normal condition
75%–99% or the cells differentiate into granular GABAergic
neurons, with the rest forming periglomular neurons expressing
either tyrosine hydroxylase or GABA. The control of
proliferation and    mobilisation   of   these   cells   may   be
dopaminergic as both MPTP and 6-OHDA mediated dopamine
depletion reportedly decreases proliferation in this zone (Zhao
et al., 2008).
   An additional source of endogenous source of new
dopaminergic neurons may be described presence of tyrosine
hydroxylase positive cell bodies in the striatum, which increase
in quantity with dopaminergic denervation. As yet there are no
imminent therapeutic strategies heading towards the clinic that
manipulate these endogenous systems but their potential is
waiting to be harnessed. Therapeutic strategies to increase
striatal dopamine could involve recruiting newly produced
neurons in the SVZ and encouraging them to migrate into the
striatum and differentiate into dopaminergic neurones or to
stimulate cells resident in the striatum. In order for this to be
achieved understanding more about these two processes of
neurogenesis and phenotypic switching in the striatum is
necessary, determining the intrinsic or extrinsic factors


                               56
responsible may provide an alternative set of mechanisms that
could be utilized to treat PD (Lane et al., 2008).
Graft standardization:
   Grafting methods for the past 20 years have differed in
everything from procurement process to tissue composition to
implantation technique. To add even more variability, multiple
donors are needed to create a graft large enough to carry some
promise of efficacy. This, undoubtedly, plays an important role
in determining survival, growth, and integration of the
transplant (Morizane et al., 2009).
   As stem cells can theoretically provide an endless source of
quality consistent neurons, standardization of the transplant
tissue will enhance the reliability of the procedure and its
results. One promising study has shown that implantation of
undifferentiated   human     neural    stem cells (hNSCs)       in
Parkinsonian    primate    brains     can   restore   functionality.
Furthermore, the repair process not only reestablishes the gross
anatomical structure of the organ but does so with appropriate
proportions of neuron types. Guided by signals of the
microenvironment of the damaged brain, uncommitted hNSCs
are induced to differentiate into dopaminergic neurons, as well
as other cells that mediate neuroprotection (Lane et al., 2008).
Patient standardization:
   With standardization of transplant material, patients must
likewise be evaluated for variables in their presentation of the
disease. Specifically, the distribution of the damaged neurons


                                57
should be taken into account before and after graft implantation.
In earlier studies, patient selections overlooked the preoperative
magnitude of the lesions, making it difficult to evaluate the
extent of the graft incorporation. Similarly, it was also unknown
whether continued postoperative degeneration of non grafted
regions would affect clinical response. Conversely, patients with
little or no postoperative damage showed the best functional
outcome. Because the decline of dopaminergic cells in areas
outside the nigrostriatal region seems to arise as PD progresses
to a more severe state, it may be that implantation during earlier
stages will exhibit a higher rate of success. This is mirrored in
the survival of transplanted tissue, which survives and integrates
better in younger patients. The reasons why this occurs have yet
to be fully determined, but it is known that neural growth factors
are expressed more in younger brains (Morizane et al., 2009).




                               58
Stem cell therapy in stroke
   Stroke is defined as an abrupt focal loss of brain function
resulting from interference with the blood supply to part of the
central nervous system. It is one of the major causes of death
and disability among the adult population in the world. In spite
of the extensive research in the field of stroke biology, there is
little effective treatment for a completed stroke. Most strokes
fall into two main categories: ischemic (80%) or hemorrhagic
(20%) (Caplan, 2011).
The biology of cellular transplantation:
   The transplantation of human neuronal cells is an approach
to reducing the functional deficits caused by CNS disease or
injury. Several investigators have evaluated the effects of
transplanted fetal tissue, rat striatum, or cellular implants into
small animal stroke models for the most part, clinical trial
designs using primary human fetal tissue into patients with
neurodegenerative diseases have lessened. The widespread
clinical use of primary human tissue is likely to be limited due
to the ethical and technical difficulties in obtaining large
quantities of fetal neurons at the same time; much effort has
been devoted to developing alternate sources of human neurons
for use in transplantation (Kondziolka and Wechsler, 2008).
   When transplanted, these neuronal cells survived, extended
processes, expressed neurotransmitters,      formed functional
synapses, and integrated with the host. During the retinoic acid


                               59
induction process, significant changes were seen in the neuron
precursor cells that resulted in the loss of neuroepithelial
markers and the appearance of neuronal markers. The final cell
product was a ≥ 95% pure population of human neuronal cells
that appeared virtually indistin-guishable from terminally
differentiated, post mitotic neurons. The cells were capable of
differentiation to express a variety of neuronal markers
characteristic of mature neurons, including all 3 neurofilament
proteins (L, M, and H); microtubule associated protein 2, the
somatic/dendritic protein, the axonal protein. Thus, the neuronal
phenotype made these cells a promising candidate for
replacement in patients with CNS disorders, as a virtually
unlimited supply of pure, post mitotic, and differentiated human
neuronal cells (Lindvall and Kokaia, 2010).
    The concept of restoring function after a stroke by
transplanting human neuronal cells into the brain was conceived
in the mid 1990s. Research conducted in a rat model of transient
focal cerebral ischemia demonstrated that transplantation of
fetal tissue restored both cognitive and motor         functions.
Ischemic stroke leads to the death of multiple neuronal types
and astrocytes, oligodendrocytes, and endothelial cells in the
cortex and subcortical regions. Stem cell based therapy could be
used to restore damaged neural circuitry by transplanting stem
cell derived neuron precursors/neuroblasts. Also, compounds
could be infused that would promote neurogenesis from
endogenous SVZ stem/progenitor cells, or stem cells could be


                               60
injected systemically for neuroprotection and modulation of
inflammation (Kondziolka and Wechsler, 2008).
   Behavioral testing was conducted using a passive avoidance
learning and retention task and a motor asymmetry measure.
Animals that received transplants of neurons and treatment with
cyclosporine showed        amelioration     of   ischemia     induced
behavioral deficits throughout the 6 month observation period.
They demonstrated complete recovery in the passive avoidance
test, as well as normalization of motor function in the elevated
body swing test. In comparison, control groups receiving
transplants of rat fetal cerebellar cells, medium alone, or
cyclosporine failed to show significant behavioral improvement.
Subsequent studies have shown that these cells released glial
derived neurotrophic factor after transplantation into ischemic
rats (figure 18) (Lindvall and Kokaia, 2010).




Figure (18): Stem cell based therapies for stroke (Lindvall and Kokaia,
2010).



                                  61
Embryonic stem cells:
   Animal models have demonstrated that ESCs, when
transplanted into adult hosts, differentiate and develop into cells
and tissues and thus may be applicable for treating a variety of
conditions, including Parkinson’s disease, multiple sclerosis,
spinal injuries, stroke, and cancer. Transplanted ESCs are
exposed to immune reactions similar to those acting on organ
transplants; hence, immunosuppression of the recipient is
generally required. It is possible, however, to obtain ESCs that
are genetically identical to the patient’s own cells using
therapeutic cloning techniques (Kalluri and Dempsey, 2008).
   Several studies showed that these cells are able to migrate in
response to damage, using MRI, that ESCs that were implanted
into the healthy hemisphere of rat brains 2 weeks after focal
cerebral ischemia (FCI), migrated along the corpus callosum to
the ventricular walls, and populated en masse at the border zone
of the damaged brain tissue (i.e., the hemisphere opposite to the
implantation sites). Another study showed that undifferentiated
ESCs xenotransplanted into the rat brain at the hemisphere
opposite to the ischemic injury migrated along the corpus
callosum toward the damaged tissue and differentiated into
neurons at the border zone of the lesion. In the homologous
mouse brain, the same murine ESCs did not migrate, but
produced highly malignant teratocarcinomas at the site of
implantation,    independent      of    whether     they     were
predifferentiated in vitro to NPCs. These results imply that


                                62
ESCs might migrate to the damaged site. However, the
production of teratocarcinoma raises concerns about the safety
of ESC transplantation in patients with stroke (Battler and Leor,
2006).
Adult neural stem cells:
         During the last century, the dogma existed that the adult
CNS was incapable of generating new neurons (neurogenesis).
Over the past decades, convincing evidence emerged that
neurogenesis in the adult CNS is a continuous physiological
process. Neurogenesis          is present      in   two regions:       the
subventricular zone (SVZ) and the subgranular zone of the
dentate gyrus (figure 19) (Kalluri and Dempsey, 2008).




Figure (19): Schematic drawings showing ischemia induced damage in the
cortex and neural stem cell proliferation in the subventricular zone and
dentate gyrus; these hypothetical sections of brain illustrate the ischemic
tissue and the neurogenic regions. A: Drawing showing the subventricular
zone of the lateral ventricles. B: Drawing showing the dentate gyrus in the
hippocampus. Note the migration of cells from the subventricular zone
toward the infarcted area. Red dots represent proliferating and/or migrating
neural stem and/or progenitor cells (Kalluri and Dempsey, 2008).




                                    63
Additionally, other studies also indicated the existence of
NSCs in other regions of the CNS, namely the striatum, spinal
cord and neocortex. SVZ and dentate gyrus derived NSCs are
characterized by long term, self renewal capacity and
multipotency. Adult SVZ and dentate gyrus derived NSCs
persist throughout the life span of mammals including humans.
It is important to note that neurogenesis occurs in a
physiological mode or is exogenously modulated by external
signals or pathophysiological processes. External global
stimulants such as enriched environment, physical activity and
stress or application of defined molecules such as fibroblast
growth factor-2, vascular endothelial growth factor (VEGF),
brain derived neurotrophic factor (BDNF) and erythropoietin
differentially modulate adult neurogenesis. Finally, CNS disease
conditions such as seizures and traumatic brain injury
represented by respective animal models induce neurogenesis
(Panchision, 2006).
Role of neural stem cells:
Proliferation:
   Neural stem cell proliferation involves the sequential
activation of several cell cycle dependent enzymes and proteins
to initiate either symmetrical (2 stem cells) or asymmetrical cell
division (1 stem and 1 progenitor cell). Erroneous activation of
cell cycle enzymes in differentiated cells like neurons, however,
can lead to either apoptotic death of the neurons or to the
formation of cancer cells. A plethora of growth factors are


                               64
expressed by the ischemic tissue, which may be responsible for
the ischemia induced neurogenesis. The increase in ischemia
induced neurogenesis could therefore be due to the upregulation
of growth factor content or their receptor expression (Hass et
al., 2005).
   In addition, not all growth factors are stimulatory in
function. Although a variety of growth and trophic factors are
upregulated following ischemic injury, some are stimulators of
neural progenitor proliferation, whereas others block the self
renewal of cells. It is important to note that ischemia induced
migration of neuroblasts has been shown to be due to the
expression of monocyte chemoattractant protein (MCP-1),
which can attract the progenitor cells away from the neurogenic
niche, blocking their proliferation. Likewise, inhibition of
growth factor activity by its binding proteins may also interfere
with proliferation of cells. Interestingly, insulin like growth
factor–I (IGF-I) can stimulate the proliferation of progenitor
cells only in the presence of mitogens like fibroblast growth
factor–2 (FGF-2) and promotes differentiation following
mitogen (FGF-2) withdrawal (figure 20) (Kalluri and Dempsey,
2008).




                               65
Figure (20): Schematic drawing showing proliferation and differentiation
of neural stem cells. These cells proliferate in response to mitogens and
differentiate into neurons, astrocytes, and oligodendrocytes on exposure to
various growth factors (Kalluri and Dempsey, 2008).


Migration:
   Several studies have shown that ischemia induces the
migration of neuroblasts into the striatum and cerebral cortex.
An enriched environment, however, increased the stroke
induced neurogenesis in the hippocampus, but decreased the cell
genesis and migration of neuroblasts into striatum. In addition,
erythropoietin has also been shown to be involved in the
proliferation of progenitor cells in the subventricular zone.
Hence, it appears that proliferation and migration of cells may
be interrelated, in the sense that they are elements of the same
process (Son et al., 2006).



                                   66
It is thought that chemokines proteins released at the site of
injury by the inflammatory cells induce migration of the cells.
The expression of chemokine receptors on neural stem cells is
important for an efficient response to chemokines. The
expression of chemokine receptors is regulated by retinoic acid,
and that animals fed on a diet lacking retinoic acid have
decreased numbers of double cortin positive cells. Retinoic acid
has been shown to induce the differentiation of neurons in vitro.
Hence, it appears that lack of retinoic acid may have decreased
the neuronal differentiation and/ or the expression of chemokine
receptors, which results in decreased numbers of neuroblasts
(immature neuronal cells expressing double cortin) migrating
out of the neurogenic niche (figure 21) (Kondziolka and
Wechsler, 2008).




                               67
Figure (21): Schematic drawing showing ischemia induced neurogenesis.
Ischemia increases the expression of IGF-I, FGF- 2, TGFb1, and MCP-1.
The IGF-I and FGF-2 enhance the proliferation of cells, whereas MCP-1
increases the migration of cells away from neurogenic regions toward the
ischemic tissue. The stem cells leaving the neurogenic regions exit the cell
cycle, during which time TGFb1 promotes the differentiation of stem cells
into neuroblasts (migrating immature neurons). Alternately, if proliferating
cells in the neurogenic region are exposed to TGFb1, it will inhibit the
process, thus slowing down the proliferation of cells (Kondziolka and
Wechsler, 2008).


Differentiation:
    The process of generating specialized cells from neural stem
cells is called differentiation. Neural stem cells can be
differentiated into neurons, astrocytes, and oligodendrocytes;
this involves interplay between intrinsic cellular programs and
extrinsic cues like growth factors provided by the surrounding
environment. Although neural progenitor cells proliferate in
response to ischemia, several immature neurons (double cortin
positive cells) were shown to be migrating toward striatum and
cortex (Battler and Leor, 2006).


                                    68
Although IGF-I induces the proliferation and differentiation
of progenitor cells after ischemia, most of the cells that are
migrating toward the injury site are neuroblasts (immature
neurons) but not oligodendrocytes. Although speculative, a
neuronal differentiation factor like transforming growth factor
b1 (TGFb1), which is upregulated after ischemia, may be
responsible for the formation of neuroblasts during the post
ischemic neurogenesis. The differential response of IGF-I
toward proliferation and differentiation was shown to be
regulated by a mitogen activated protein kinase pathway.
Because matrix metalloproteinase (MMPs) play an important
role in both extracellular matrix (ECM) digestion and IGF
regulation, it is crucial to understand their direct (ECM
digestion) and indirect (IGF metabolism) effects on progenitor
cell differentiation. Hence it is possible that after stroke, cells
proliferate in response to mitogens and growth factors, some of
which exit the cell cycle due to the chemokine mediated
migration out of the neurogenic area, and differentiate into
neuroblasts in response to differentiation promoting factors such
as TGFb1. Thus, the final outcome depends on the spatial and
temporal expression of these factors following ischemia (Son et
al., 2006).
Potential     mechanism   involved    in stem     cell mediated
recovery after stroke:
   Initial transplantation studies were focused on the potential
of NSCs to replace lost circuitry. Transplanted neural progenitor


                                69
cell (NPCs) in a rat model of global ischemia have been
reported to express synaptic proteins post transplantation.
However, only limited evidence demonstrates that transplanted
cells are able to sustain CNS repair through massive cell
replacement, especially to the extent that might be required after
stroke. Regardless of the characteristics of the experimental
disease,     functional   recovery       achieved    through   NPC
transplantation does not correlate with absolute numbers of
transplant    derived,    newly        generated,   and   terminally
differentiated neural cells (Emerson et al., 2008).
Stem cell induced neuroprotection:
   Transplanted stem cells can provide neuroprotection and
reduce host cell death in the post stroke brain. Most authors
have reported functional recovery and a reduction in lesion size
when cells are transplanted within the first 24-48 hours after
stroke. The short timeframe in which NPCs affect recovery
cannot be explained by the regeneration of new neurons and
synapses, suggesting an important role for neuroprotection in
enhancing recovery. In fact, NPCs are known to exert direct
neuroprotection through the neutralization of free radicals,
inflammatory cytokines, excitotoxins, lipases, peroxidases, and
other toxic metabolites that are released following an ischemic
event (Ourednik et al., 2005).
   The neuroprotective effects of transplanted NPCs are usually
accompanied by increased in vivo bioavailability of main
neurotrophins such as nerve growth factor, brain derived


                                  70
neurotrophic factor (BDNF), ciliary neurotrophic factor,
vascular endothelial growth factor (VEGF), fibroblast growth
factor, and    glial   derived    neurotrophic factor (GDNF).
Alternatively, one could argue that the cells did adhere to the
endothelium in the affected brain area and exerted their effect
through the secretion of GDNF but did not eventually engraft. A
different aspect of neuroprotection would be an effect on
endogenous     neurogenesis       after      stroke.   Endogenous
neurogenesis is increased after a stroke. Its exact function has
yet to be determined, but it may signify a natural repair
mechanism of the brain, which could be enhanced by
transplanted cells (Guzman et al., 2008).
Stem cell induced angiogenesis:
   Transplanted    NPCs     can       also   enhance   endogenous
angiogenesis. Increased vascularization in the penumbra within
a few days after stroke is associated with spontaneous functional
recovery. As early as 3 days after ischemic injury, new blood
vessels are observed in the stroke affected penumbra, and
growth continues to increase for at least 21 days. Transplanted
cell induced blood vessel formation has been reported with
BMSCs, NSCs, and cells from human cord blood or peripheral
blood. The ability of transplanted cells to increase endogenous
levels of angiogenic factors and chemoattractant factors (for
example, SDF-1) induces the proliferation of existing vascular
endothelial cells (angiogenesis) and mobilization and homing of




                                 71
endogenous endothelial progenitors (vasculogenesis) (Shyu et
al., 2006).
   The direct injection of recombinant stromal derived factor–1
(SDF-1) into the stroke affected rat brain resulted in the
increased recruitment of BMSCs and increased vascular density
in the ischemic brain. A similar effect was achieved by
treatment of stroke affected rats with granulocyte colony
stimulating factor (Ourednik et al., 2005).
Stem cell induced immunomodulation:
   In addition to       neuroprotection and enhancement       of
angiogenesis, transplanted NPCscan also decrease post
ischemic inflammatory damage. A landmark study highlights
the remarkable ability of transplanted NPCs to promote
neuroprotection through an immunomodulatory strategy, once
within inflamed CNS areas, systemically injected NPCs persist
around the perivascular space where reactive astrocytes,
inflamed endothelial cells, and bloodborne infiltrating T cells
coreside. Adult NPCs can promote neuroprotection by releasing
antiinflammatory        chemokines     and    by      expressing
immunomodulatory molecules (Pluchino et al., 2005).
   In animal models of stroke, decreased infiltration of
mononuclear cells has been demonstrated at the lesion borders
of ischemic areas in the CNS where NPCs accumulate. Working
under the paradigm of bidirectional communication between
transplanted    cells    and    the    inflammatory    ischemic
microenvironment, one group has attempted to combine NPC


                               72
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology
Thesis section: Restorative neurology

Más contenido relacionado

La actualidad más candente

Stem cells in CNS disorders
Stem cells in CNS disordersStem cells in CNS disorders
Stem cells in CNS disordersdevang084
 
Stem Cells in the Treatment of Alzheimer's Disease
Stem Cells in the Treatment of Alzheimer's DiseaseStem Cells in the Treatment of Alzheimer's Disease
Stem Cells in the Treatment of Alzheimer's DiseaseNatasha Delgado
 
Stem Cell Treatment At Tiantan Puhua Hospital 4
Stem  Cell  Treatment At  Tiantan  Puhua  Hospital  4Stem  Cell  Treatment At  Tiantan  Puhua  Hospital  4
Stem Cell Treatment At Tiantan Puhua Hospital 4randyrobinsonpuhua
 
Neuronal stem cellfi
Neuronal stem cellfiNeuronal stem cellfi
Neuronal stem cellfiDUVASU
 
ppt- paper 303.pptx
ppt- paper 303.pptxppt- paper 303.pptx
ppt- paper 303.pptxManjunathAT8
 
Stem cells and regenerative medicine
Stem cells and regenerative medicineStem cells and regenerative medicine
Stem cells and regenerative medicineJibanjyotiDas2
 
Stem cells self renewal and lineage
Stem cells self renewal and lineageStem cells self renewal and lineage
Stem cells self renewal and lineageExicanLifeSciences
 
Stem cells in regenrative therapy
Stem cells in regenrative therapyStem cells in regenrative therapy
Stem cells in regenrative therapyRaghavendra Raghu
 
EXTRACELLULAR VESICLES IN CANCER
EXTRACELLULAR VESICLES IN CANCEREXTRACELLULAR VESICLES IN CANCER
EXTRACELLULAR VESICLES IN CANCERAniket Vaidya
 
Exosomes -power point presentation
 Exosomes -power point presentation Exosomes -power point presentation
Exosomes -power point presentationmeghanamegha23
 
Exosomes - Diagnostics and Therapeutics
Exosomes - Diagnostics and TherapeuticsExosomes - Diagnostics and Therapeutics
Exosomes - Diagnostics and TherapeuticsSumedhaBobade
 
Modulation_of_autophagy_as_a_therapeutic
Modulation_of_autophagy_as_a_therapeuticModulation_of_autophagy_as_a_therapeutic
Modulation_of_autophagy_as_a_therapeuticYildiz Kelahmetoglu
 
Review paper ms 1 revised2
Review paper ms 1 revised2Review paper ms 1 revised2
Review paper ms 1 revised2lysanderborrero
 

La actualidad más candente (20)

Stem cells in CNS disorders
Stem cells in CNS disordersStem cells in CNS disorders
Stem cells in CNS disorders
 
Stem Cells in the Treatment of Alzheimer's Disease
Stem Cells in the Treatment of Alzheimer's DiseaseStem Cells in the Treatment of Alzheimer's Disease
Stem Cells in the Treatment of Alzheimer's Disease
 
Cell
CellCell
Cell
 
Stem Cell Treatment At Tiantan Puhua Hospital 4
Stem  Cell  Treatment At  Tiantan  Puhua  Hospital  4Stem  Cell  Treatment At  Tiantan  Puhua  Hospital  4
Stem Cell Treatment At Tiantan Puhua Hospital 4
 
Stem cell in Neurosurgery
Stem cell in NeurosurgeryStem cell in Neurosurgery
Stem cell in Neurosurgery
 
Neuronal stem cellfi
Neuronal stem cellfiNeuronal stem cellfi
Neuronal stem cellfi
 
STEMCELL
STEMCELLSTEMCELL
STEMCELL
 
Stem cells & Neurodegenerative diseases And Some clinical CNS
Stem cells & Neurodegenerative diseases And Some clinical CNSStem cells & Neurodegenerative diseases And Some clinical CNS
Stem cells & Neurodegenerative diseases And Some clinical CNS
 
ppt- paper 303.pptx
ppt- paper 303.pptxppt- paper 303.pptx
ppt- paper 303.pptx
 
Stange1965
Stange1965Stange1965
Stange1965
 
Stem cells and regenerative medicine
Stem cells and regenerative medicineStem cells and regenerative medicine
Stem cells and regenerative medicine
 
Stem cells self renewal and lineage
Stem cells self renewal and lineageStem cells self renewal and lineage
Stem cells self renewal and lineage
 
Review Paper Natasha
Review Paper NatashaReview Paper Natasha
Review Paper Natasha
 
Stem cells in regenrative therapy
Stem cells in regenrative therapyStem cells in regenrative therapy
Stem cells in regenrative therapy
 
EXTRACELLULAR VESICLES IN CANCER
EXTRACELLULAR VESICLES IN CANCEREXTRACELLULAR VESICLES IN CANCER
EXTRACELLULAR VESICLES IN CANCER
 
Exosomes -power point presentation
 Exosomes -power point presentation Exosomes -power point presentation
Exosomes -power point presentation
 
Exosomes - Diagnostics and Therapeutics
Exosomes - Diagnostics and TherapeuticsExosomes - Diagnostics and Therapeutics
Exosomes - Diagnostics and Therapeutics
 
Modulation_of_autophagy_as_a_therapeutic
Modulation_of_autophagy_as_a_therapeuticModulation_of_autophagy_as_a_therapeutic
Modulation_of_autophagy_as_a_therapeutic
 
Review paper ms 1 revised2
Review paper ms 1 revised2Review paper ms 1 revised2
Review paper ms 1 revised2
 
Tissue Engineering slides
Tissue Engineering slidesTissue Engineering slides
Tissue Engineering slides
 

Similar a Thesis section: Restorative neurology

Stem cells are biological cells found in all multicellular organisms
Stem cells are biological cells found in all multicellular organismsStem cells are biological cells found in all multicellular organisms
Stem cells are biological cells found in all multicellular organismsytgyuy
 
A Review Paper On Stem Cells
A Review Paper On Stem CellsA Review Paper On Stem Cells
A Review Paper On Stem CellsErica Thompson
 
Christian drapeau theory_of_regeneration
Christian drapeau theory_of_regenerationChristian drapeau theory_of_regeneration
Christian drapeau theory_of_regenerationbestwebsite2008
 
Stem cell primordial_food_excerpt
Stem cell primordial_food_excerptStem cell primordial_food_excerpt
Stem cell primordial_food_excerptbestwebsite2008
 
Stem cells #scichallenge2017
Stem cells  #scichallenge2017Stem cells  #scichallenge2017
Stem cells #scichallenge2017Vait Erin
 
The ISSCR is an independent, nonprofit organization providin.docx
The ISSCR is an independent, nonprofit organization providin.docxThe ISSCR is an independent, nonprofit organization providin.docx
The ISSCR is an independent, nonprofit organization providin.docxoreo10
 
Stem cell Research
Stem cell ResearchStem cell Research
Stem cell ResearchSatarupa Deb
 
STEM CELLS IN PERIODONTICS
STEM CELLS IN PERIODONTICSSTEM CELLS IN PERIODONTICS
STEM CELLS IN PERIODONTICSPrajakta Phadke
 
The therapeutic potential of stem cells from adults
The therapeutic potential of stem cells from adultsThe therapeutic potential of stem cells from adults
The therapeutic potential of stem cells from adultsbestwebsite2008
 
The therapeutic potential of stem cells from adults
The therapeutic potential of stem cells from adultsThe therapeutic potential of stem cells from adults
The therapeutic potential of stem cells from adultsbestwebsite2008
 
Primordial foodv0105v1 new
Primordial foodv0105v1 newPrimordial foodv0105v1 new
Primordial foodv0105v1 newandavar
 
Stem cell tisue final version
Stem cell tisue final versionStem cell tisue final version
Stem cell tisue final versionHovo Khachikyan
 
Stem cell tisue final version
Stem cell tisue final versionStem cell tisue final version
Stem cell tisue final versionHovo Khachikyan
 

Similar a Thesis section: Restorative neurology (20)

Regenerative medicine
Regenerative medicineRegenerative medicine
Regenerative medicine
 
Stem cells are biological cells found in all multicellular organisms
Stem cells are biological cells found in all multicellular organismsStem cells are biological cells found in all multicellular organisms
Stem cells are biological cells found in all multicellular organisms
 
Stem cells
Stem cellsStem cells
Stem cells
 
A Review Paper On Stem Cells
A Review Paper On Stem CellsA Review Paper On Stem Cells
A Review Paper On Stem Cells
 
Stem Cell
Stem CellStem Cell
Stem Cell
 
Stem Cell
Stem CellStem Cell
Stem Cell
 
Christian drapeau theory_of_regeneration
Christian drapeau theory_of_regenerationChristian drapeau theory_of_regeneration
Christian drapeau theory_of_regeneration
 
Stem cell primordial_food_excerpt
Stem cell primordial_food_excerptStem cell primordial_food_excerpt
Stem cell primordial_food_excerpt
 
Stem cells #scichallenge2017
Stem cells  #scichallenge2017Stem cells  #scichallenge2017
Stem cells #scichallenge2017
 
The ISSCR is an independent, nonprofit organization providin.docx
The ISSCR is an independent, nonprofit organization providin.docxThe ISSCR is an independent, nonprofit organization providin.docx
The ISSCR is an independent, nonprofit organization providin.docx
 
Stem cell Research
Stem cell ResearchStem cell Research
Stem cell Research
 
Regenerative medicine 2006
Regenerative medicine 2006Regenerative medicine 2006
Regenerative medicine 2006
 
STEM CELLS IN PERIODONTICS
STEM CELLS IN PERIODONTICSSTEM CELLS IN PERIODONTICS
STEM CELLS IN PERIODONTICS
 
Stem cells
Stem cellsStem cells
Stem cells
 
The therapeutic potential of stem cells from adults
The therapeutic potential of stem cells from adultsThe therapeutic potential of stem cells from adults
The therapeutic potential of stem cells from adults
 
The therapeutic potential of stem cells from adults
The therapeutic potential of stem cells from adultsThe therapeutic potential of stem cells from adults
The therapeutic potential of stem cells from adults
 
Primordial foodv0105v1 new
Primordial foodv0105v1 newPrimordial foodv0105v1 new
Primordial foodv0105v1 new
 
Stem cell tisue final version
Stem cell tisue final versionStem cell tisue final version
Stem cell tisue final version
 
Stem cell tisue final version
Stem cell tisue final versionStem cell tisue final version
Stem cell tisue final version
 
Name .docx
Name                                                             .docxName                                                             .docx
Name .docx
 

Más de Professor Yasser Metwally

The Snake, the Scorpion, the turtle in Egypt
The Snake, the Scorpion, the turtle in EgyptThe Snake, the Scorpion, the turtle in Egypt
The Snake, the Scorpion, the turtle in EgyptProfessor Yasser Metwally
 
Radiological pathology of epileptic disorders
Radiological pathology of epileptic disordersRadiological pathology of epileptic disorders
Radiological pathology of epileptic disordersProfessor Yasser Metwally
 
Radiological pathology of cerebrovascular disorders
Radiological pathology of cerebrovascular disordersRadiological pathology of cerebrovascular disorders
Radiological pathology of cerebrovascular disordersProfessor Yasser Metwally
 
Radiological pathology of spontaneous cerebral hemorrhage
Radiological pathology of spontaneous cerebral hemorrhageRadiological pathology of spontaneous cerebral hemorrhage
Radiological pathology of spontaneous cerebral hemorrhageProfessor Yasser Metwally
 
Radiological pathology of cerebral amyloid angiography
Radiological pathology of cerebral amyloid angiographyRadiological pathology of cerebral amyloid angiography
Radiological pathology of cerebral amyloid angiographyProfessor Yasser Metwally
 
Radiological pathology of cerebral microbleeds
Radiological pathology of cerebral microbleedsRadiological pathology of cerebral microbleeds
Radiological pathology of cerebral microbleedsProfessor Yasser Metwally
 
Issues in radiological pathology: Radiological pathology of watershed infarct...
Issues in radiological pathology: Radiological pathology of watershed infarct...Issues in radiological pathology: Radiological pathology of watershed infarct...
Issues in radiological pathology: Radiological pathology of watershed infarct...Professor Yasser Metwally
 
Radiological pathology of cortical laminar necrosis
Radiological pathology of cortical laminar necrosisRadiological pathology of cortical laminar necrosis
Radiological pathology of cortical laminar necrosisProfessor Yasser Metwally
 

Más de Professor Yasser Metwally (20)

The Egyptian Zoo in Cairo 2015
The Egyptian Zoo in Cairo 2015The Egyptian Zoo in Cairo 2015
The Egyptian Zoo in Cairo 2015
 
End of the great nile river in Ras Elbar
End of the great nile river in Ras ElbarEnd of the great nile river in Ras Elbar
End of the great nile river in Ras Elbar
 
The Lion and The tiger in Egypt
The Lion and The tiger in EgyptThe Lion and The tiger in Egypt
The Lion and The tiger in Egypt
 
The monkeys in Egypt
The monkeys in EgyptThe monkeys in Egypt
The monkeys in Egypt
 
The Snake, the Scorpion, the turtle in Egypt
The Snake, the Scorpion, the turtle in EgyptThe Snake, the Scorpion, the turtle in Egypt
The Snake, the Scorpion, the turtle in Egypt
 
The Egyptian Parrot
The Egyptian ParrotThe Egyptian Parrot
The Egyptian Parrot
 
The Egyptian Deer
The Egyptian DeerThe Egyptian Deer
The Egyptian Deer
 
The Egyptian Pelican
The Egyptian PelicanThe Egyptian Pelican
The Egyptian Pelican
 
The Flamingo bird in Egypt
The Flamingo bird in EgyptThe Flamingo bird in Egypt
The Flamingo bird in Egypt
 
Egyptian Cats
Egyptian CatsEgyptian Cats
Egyptian Cats
 
Radiological pathology of epileptic disorders
Radiological pathology of epileptic disordersRadiological pathology of epileptic disorders
Radiological pathology of epileptic disorders
 
Radiological pathology of cerebrovascular disorders
Radiological pathology of cerebrovascular disordersRadiological pathology of cerebrovascular disorders
Radiological pathology of cerebrovascular disorders
 
Radiological pathology of spontaneous cerebral hemorrhage
Radiological pathology of spontaneous cerebral hemorrhageRadiological pathology of spontaneous cerebral hemorrhage
Radiological pathology of spontaneous cerebral hemorrhage
 
Radiological pathology of cerebral amyloid angiography
Radiological pathology of cerebral amyloid angiographyRadiological pathology of cerebral amyloid angiography
Radiological pathology of cerebral amyloid angiography
 
Radiological pathology of cerebral microbleeds
Radiological pathology of cerebral microbleedsRadiological pathology of cerebral microbleeds
Radiological pathology of cerebral microbleeds
 
The Egyptian Zoo in Cairo
The Egyptian Zoo in CairoThe Egyptian Zoo in Cairo
The Egyptian Zoo in Cairo
 
Progressive multifocal leukoencephalopathy
Progressive multifocal leukoencephalopathyProgressive multifocal leukoencephalopathy
Progressive multifocal leukoencephalopathy
 
Progressive multifocal leukoencephalopathy
Progressive multifocal leukoencephalopathyProgressive multifocal leukoencephalopathy
Progressive multifocal leukoencephalopathy
 
Issues in radiological pathology: Radiological pathology of watershed infarct...
Issues in radiological pathology: Radiological pathology of watershed infarct...Issues in radiological pathology: Radiological pathology of watershed infarct...
Issues in radiological pathology: Radiological pathology of watershed infarct...
 
Radiological pathology of cortical laminar necrosis
Radiological pathology of cortical laminar necrosisRadiological pathology of cortical laminar necrosis
Radiological pathology of cortical laminar necrosis
 

Último

Bangalore Call Girls Nelamangala Number 7001035870 Meetin With Bangalore Esc...
Bangalore Call Girls Nelamangala Number 7001035870  Meetin With Bangalore Esc...Bangalore Call Girls Nelamangala Number 7001035870  Meetin With Bangalore Esc...
Bangalore Call Girls Nelamangala Number 7001035870 Meetin With Bangalore Esc...narwatsonia7
 
Call Girls Bhubaneswar Just Call 9907093804 Top Class Call Girl Service Avail...
Call Girls Bhubaneswar Just Call 9907093804 Top Class Call Girl Service Avail...Call Girls Bhubaneswar Just Call 9907093804 Top Class Call Girl Service Avail...
Call Girls Bhubaneswar Just Call 9907093804 Top Class Call Girl Service Avail...Dipal Arora
 
Call Girls Kochi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Kochi Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Kochi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Kochi Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
(Rocky) Jaipur Call Girl - 09521753030 Escorts Service 50% Off with Cash ON D...
(Rocky) Jaipur Call Girl - 09521753030 Escorts Service 50% Off with Cash ON D...(Rocky) Jaipur Call Girl - 09521753030 Escorts Service 50% Off with Cash ON D...
(Rocky) Jaipur Call Girl - 09521753030 Escorts Service 50% Off with Cash ON D...indiancallgirl4rent
 
The Most Attractive Hyderabad Call Girls Kothapet 𖠋 6297143586 𖠋 Will You Mis...
The Most Attractive Hyderabad Call Girls Kothapet 𖠋 6297143586 𖠋 Will You Mis...The Most Attractive Hyderabad Call Girls Kothapet 𖠋 6297143586 𖠋 Will You Mis...
The Most Attractive Hyderabad Call Girls Kothapet 𖠋 6297143586 𖠋 Will You Mis...chandars293
 
Call Girls Cuttack Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Cuttack Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Cuttack Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Cuttack Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Top Rated Bangalore Call Girls Mg Road ⟟ 9332606886 ⟟ Call Me For Genuine Se...
Top Rated Bangalore Call Girls Mg Road ⟟  9332606886 ⟟ Call Me For Genuine Se...Top Rated Bangalore Call Girls Mg Road ⟟  9332606886 ⟟ Call Me For Genuine Se...
Top Rated Bangalore Call Girls Mg Road ⟟ 9332606886 ⟟ Call Me For Genuine Se...narwatsonia7
 
Call Girls Gwalior Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Gwalior Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Gwalior Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Gwalior Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
VIP Hyderabad Call Girls Bahadurpally 7877925207 ₹5000 To 25K With AC Room 💚😋
VIP Hyderabad Call Girls Bahadurpally 7877925207 ₹5000 To 25K With AC Room 💚😋VIP Hyderabad Call Girls Bahadurpally 7877925207 ₹5000 To 25K With AC Room 💚😋
VIP Hyderabad Call Girls Bahadurpally 7877925207 ₹5000 To 25K With AC Room 💚😋TANUJA PANDEY
 
Call Girls Jabalpur Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Jabalpur Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Jabalpur Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Jabalpur Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Call Girls Bangalore Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Bangalore Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Bangalore Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Bangalore Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Call Girls Faridabad Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Faridabad Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Faridabad Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Faridabad Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Call Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore Escorts
Call Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore EscortsCall Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore Escorts
Call Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore Escortsvidya singh
 
Russian Escorts Girls Nehru Place ZINATHI 🔝9711199012 ☪ 24/7 Call Girls Delhi
Russian Escorts Girls  Nehru Place ZINATHI 🔝9711199012 ☪ 24/7 Call Girls DelhiRussian Escorts Girls  Nehru Place ZINATHI 🔝9711199012 ☪ 24/7 Call Girls Delhi
Russian Escorts Girls Nehru Place ZINATHI 🔝9711199012 ☪ 24/7 Call Girls DelhiAlinaDevecerski
 
Call Girls Visakhapatnam Just Call 9907093804 Top Class Call Girl Service Ava...
Call Girls Visakhapatnam Just Call 9907093804 Top Class Call Girl Service Ava...Call Girls Visakhapatnam Just Call 9907093804 Top Class Call Girl Service Ava...
Call Girls Visakhapatnam Just Call 9907093804 Top Class Call Girl Service Ava...Dipal Arora
 
Book Paid Powai Call Girls Mumbai 𖠋 9930245274 𖠋Low Budget Full Independent H...
Book Paid Powai Call Girls Mumbai 𖠋 9930245274 𖠋Low Budget Full Independent H...Book Paid Powai Call Girls Mumbai 𖠋 9930245274 𖠋Low Budget Full Independent H...
Book Paid Powai Call Girls Mumbai 𖠋 9930245274 𖠋Low Budget Full Independent H...Call Girls in Nagpur High Profile
 
College Call Girls in Haridwar 9667172968 Short 4000 Night 10000 Best call gi...
College Call Girls in Haridwar 9667172968 Short 4000 Night 10000 Best call gi...College Call Girls in Haridwar 9667172968 Short 4000 Night 10000 Best call gi...
College Call Girls in Haridwar 9667172968 Short 4000 Night 10000 Best call gi...perfect solution
 
Call Girls Gwalior Just Call 8617370543 Top Class Call Girl Service Available
Call Girls Gwalior Just Call 8617370543 Top Class Call Girl Service AvailableCall Girls Gwalior Just Call 8617370543 Top Class Call Girl Service Available
Call Girls Gwalior Just Call 8617370543 Top Class Call Girl Service AvailableDipal Arora
 
(👑VVIP ISHAAN ) Russian Call Girls Service Navi Mumbai🖕9920874524🖕Independent...
(👑VVIP ISHAAN ) Russian Call Girls Service Navi Mumbai🖕9920874524🖕Independent...(👑VVIP ISHAAN ) Russian Call Girls Service Navi Mumbai🖕9920874524🖕Independent...
(👑VVIP ISHAAN ) Russian Call Girls Service Navi Mumbai🖕9920874524🖕Independent...Taniya Sharma
 
Call Girls Haridwar Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Haridwar Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Haridwar Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Haridwar Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 

Último (20)

Bangalore Call Girls Nelamangala Number 7001035870 Meetin With Bangalore Esc...
Bangalore Call Girls Nelamangala Number 7001035870  Meetin With Bangalore Esc...Bangalore Call Girls Nelamangala Number 7001035870  Meetin With Bangalore Esc...
Bangalore Call Girls Nelamangala Number 7001035870 Meetin With Bangalore Esc...
 
Call Girls Bhubaneswar Just Call 9907093804 Top Class Call Girl Service Avail...
Call Girls Bhubaneswar Just Call 9907093804 Top Class Call Girl Service Avail...Call Girls Bhubaneswar Just Call 9907093804 Top Class Call Girl Service Avail...
Call Girls Bhubaneswar Just Call 9907093804 Top Class Call Girl Service Avail...
 
Call Girls Kochi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Kochi Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Kochi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Kochi Just Call 9907093804 Top Class Call Girl Service Available
 
(Rocky) Jaipur Call Girl - 09521753030 Escorts Service 50% Off with Cash ON D...
(Rocky) Jaipur Call Girl - 09521753030 Escorts Service 50% Off with Cash ON D...(Rocky) Jaipur Call Girl - 09521753030 Escorts Service 50% Off with Cash ON D...
(Rocky) Jaipur Call Girl - 09521753030 Escorts Service 50% Off with Cash ON D...
 
The Most Attractive Hyderabad Call Girls Kothapet 𖠋 6297143586 𖠋 Will You Mis...
The Most Attractive Hyderabad Call Girls Kothapet 𖠋 6297143586 𖠋 Will You Mis...The Most Attractive Hyderabad Call Girls Kothapet 𖠋 6297143586 𖠋 Will You Mis...
The Most Attractive Hyderabad Call Girls Kothapet 𖠋 6297143586 𖠋 Will You Mis...
 
Call Girls Cuttack Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Cuttack Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Cuttack Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Cuttack Just Call 9907093804 Top Class Call Girl Service Available
 
Top Rated Bangalore Call Girls Mg Road ⟟ 9332606886 ⟟ Call Me For Genuine Se...
Top Rated Bangalore Call Girls Mg Road ⟟  9332606886 ⟟ Call Me For Genuine Se...Top Rated Bangalore Call Girls Mg Road ⟟  9332606886 ⟟ Call Me For Genuine Se...
Top Rated Bangalore Call Girls Mg Road ⟟ 9332606886 ⟟ Call Me For Genuine Se...
 
Call Girls Gwalior Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Gwalior Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Gwalior Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Gwalior Just Call 9907093804 Top Class Call Girl Service Available
 
VIP Hyderabad Call Girls Bahadurpally 7877925207 ₹5000 To 25K With AC Room 💚😋
VIP Hyderabad Call Girls Bahadurpally 7877925207 ₹5000 To 25K With AC Room 💚😋VIP Hyderabad Call Girls Bahadurpally 7877925207 ₹5000 To 25K With AC Room 💚😋
VIP Hyderabad Call Girls Bahadurpally 7877925207 ₹5000 To 25K With AC Room 💚😋
 
Call Girls Jabalpur Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Jabalpur Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Jabalpur Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Jabalpur Just Call 9907093804 Top Class Call Girl Service Available
 
Call Girls Bangalore Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Bangalore Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Bangalore Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Bangalore Just Call 9907093804 Top Class Call Girl Service Available
 
Call Girls Faridabad Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Faridabad Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Faridabad Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Faridabad Just Call 9907093804 Top Class Call Girl Service Available
 
Call Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore Escorts
Call Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore EscortsCall Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore Escorts
Call Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore Escorts
 
Russian Escorts Girls Nehru Place ZINATHI 🔝9711199012 ☪ 24/7 Call Girls Delhi
Russian Escorts Girls  Nehru Place ZINATHI 🔝9711199012 ☪ 24/7 Call Girls DelhiRussian Escorts Girls  Nehru Place ZINATHI 🔝9711199012 ☪ 24/7 Call Girls Delhi
Russian Escorts Girls Nehru Place ZINATHI 🔝9711199012 ☪ 24/7 Call Girls Delhi
 
Call Girls Visakhapatnam Just Call 9907093804 Top Class Call Girl Service Ava...
Call Girls Visakhapatnam Just Call 9907093804 Top Class Call Girl Service Ava...Call Girls Visakhapatnam Just Call 9907093804 Top Class Call Girl Service Ava...
Call Girls Visakhapatnam Just Call 9907093804 Top Class Call Girl Service Ava...
 
Book Paid Powai Call Girls Mumbai 𖠋 9930245274 𖠋Low Budget Full Independent H...
Book Paid Powai Call Girls Mumbai 𖠋 9930245274 𖠋Low Budget Full Independent H...Book Paid Powai Call Girls Mumbai 𖠋 9930245274 𖠋Low Budget Full Independent H...
Book Paid Powai Call Girls Mumbai 𖠋 9930245274 𖠋Low Budget Full Independent H...
 
College Call Girls in Haridwar 9667172968 Short 4000 Night 10000 Best call gi...
College Call Girls in Haridwar 9667172968 Short 4000 Night 10000 Best call gi...College Call Girls in Haridwar 9667172968 Short 4000 Night 10000 Best call gi...
College Call Girls in Haridwar 9667172968 Short 4000 Night 10000 Best call gi...
 
Call Girls Gwalior Just Call 8617370543 Top Class Call Girl Service Available
Call Girls Gwalior Just Call 8617370543 Top Class Call Girl Service AvailableCall Girls Gwalior Just Call 8617370543 Top Class Call Girl Service Available
Call Girls Gwalior Just Call 8617370543 Top Class Call Girl Service Available
 
(👑VVIP ISHAAN ) Russian Call Girls Service Navi Mumbai🖕9920874524🖕Independent...
(👑VVIP ISHAAN ) Russian Call Girls Service Navi Mumbai🖕9920874524🖕Independent...(👑VVIP ISHAAN ) Russian Call Girls Service Navi Mumbai🖕9920874524🖕Independent...
(👑VVIP ISHAAN ) Russian Call Girls Service Navi Mumbai🖕9920874524🖕Independent...
 
Call Girls Haridwar Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Haridwar Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Haridwar Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Haridwar Just Call 9907093804 Top Class Call Girl Service Available
 

Thesis section: Restorative neurology

  • 1. Restorative neurology Essay In Neuropsychiatry Submitted for partial fulfillment of Master Degree By Samy moussa Seliem M.B.B.CH Supervisors of Prof. Mohammed Yasser Metwally Professor of Neuropsychiatry Faculty of Medicine-Ain Shams University www.yassermetwally.com Prof. Naglaa Mohamed Elkhayat Professor of Neuropsychiatry Faculty of Medicine-Ain Shams University Dr. Haitham Hamdy salem Lecturer of Neuropsychiatry Faculty of Medicine-Ain Shams University Faculty of Medicine Ain Shams University 2011 i
  • 2. Contents Subject 1. Introduction and aim of the work 2. Stem cell 3. Stem cell therapy in Parkinson disease 4. Stem cell therapy in in stroke 5. Stem cell therapy in demyelinating disease 6. Stem cell therapy in in amyotrophic lateral sclerosis 7. Stem cell therapy in muscular dystrophy 8. Stem cell therapy Huntington chorea 9. Stem cell therapy for Alzheimer's Disease 10.Stem cell therapy in degenerative diseases in children 11.Stem cell therapy in retinal degeneration 12.Stem cell therapy in spinal cord injury 13.Stem cell therapy in peripheral nerve injury Summary References i
  • 3. Introduction Stem cells are unspecialized cells in the human body that are capable of becoming specialized cells, each with new specialized cell functions. The best example of a stem cell is the bone marrow stem cell that is unspecialized and able to specialize into blood cells, such as white blood cells and red blood cells, and these new cell types have special functions, such as being able to produce antibodies, act as scavengers to combat infection and transport gases. Thus one cell type stems from the other and hence the term “stem cell.” Basically, a stem cell remains uncommitted until it receives a signal to develop into a specialized cell. Stem cells have the remarkable properties of developing into a variety of cell types in the human body. They serve as a repair system by being able to divide without limit to replenish other cells. When stem cell divides, each new cell has the potential to either remain as a stem cell or become another cell type with new special functions, such as blood cells, brain cells, etc. (Bongso and Lee, 2005). Stem cells also known as progenitor cells which are cells that have not undergone differentiation to acquire specific structure or role. They have the potential to self-renew, divide and differentiate into specialized cell types. They are also, sometimes, termed ‘pluripotent’ or ‘undifferentiated’ cells i
  • 4. because they can differentiate and develop into various cell lines (Metwally, 2009). Scientists and researchers are interested in stem cells for several reasons. Although stem cells do not serve any one function, many have the capacity to serve any function after they are instructed to specialize. Every cell in the body, for example, is derived from first few stem cells formed in the early stages of embryological development. Therefore, stem cells extracted from embryos can be induced to become any desired cell type. This property makes stem cells powerful enough to regenerate damaged tissue under the right conditions (Crosta, 2010). Perhaps, the most important reason that stem cell development is so appealing to neurologists can be found in the statement “The adult human brain, in contrast to other organs such as skin and liver, lacked the capacity for self repair and regeneration” (Lin et al., 2007). The types of stem cells include: Bone marrow-derived mesenchymal stem cells (BMSCs), embryonic stem cells (ESCs), Adult (somatic) stem cells, and neural stem cells (NSCs). BMSCs also termed bone marrow stromal cells are another example of a somatic stem cell being studied for its therapeutic potential in the central nervous system (CNS) and in other tissue (Abdallah and Kassem, 2008). ii
  • 5. BMSCS generate neurotransmitter-responsive cells with electro-physiological properties similar to neurons (Diana and Gabriel, 2008). ESCs are pluripotent cells isolated from the inner cell mass of day 5-8 blastocyte with indefinite self-renewal capabilities as well as of the ability to differentiate into all cell types derived from the three embryonic germ layers. The primary therapeutic goal of ESCs research is cell replacement therapy (Aoki et al., 2007). Adult (somatic) stem cells: it has a capacity to differentiate into tissue-specific types and represent a potential source of autologus cells for transplantation therapy that eliminate immunological complications associated with allogenic donor cells as well as bypass ethical concern associated with ESCs, All types are generally characterized by their potency, or potential to differentiate into different cell types (such as skin, muscle, bone, etc) (Lin et al., 2007). Scientists discovered ways to obtain or derive stem cells from early mouse embryos more than 20 years ago. Many years of detailed study of biology of mouse stem cells led to the discovery, in 1998, of how to isolate stem cells from human embryos and grow the cells in the laboratory. These are called human embryonic stem cells. The embryos used in these studies were created for infertility purposes through in vitro fertilization iii
  • 6. procedures and when they were no longer needed for that purpose, they were donated for research with the informed consent of the donor (Ordrico et al., 2001). The concept that the adult mammalian CNS contains NSCs was first discovered from evidence of neuronal turnover in the olfactory bulb and hippocampus in the adult organism cells with more restricted neural differentiation capabilities committed to specific subpopulation lineage, have been generated from human ESCs or directly isolated from neurogenic regions of fetal and adult CNS, such as the subventricular zone, which provides neuroblasts to replenish inhibitory interneurons in the olfactory bulb (Lin et al., 2007). Stem cell differentiation must be turned on, given direction, and turned off as needed in order to properly supply the basic building blocks of tissues in different organ systems. This requirement for precise regulation applies to an even greater degree to the differentiation of neuronal progenitor cells, because effective neural function depends on establishing precise linkage and interactions between different individual neurons and classes of neurons (Metwally, 2009). Most tissue repair events in mammals are dedifferentiation independent events brought about by the activation of pre- existing stem cells or progenitor cells. By definition, a progenitor cell lies in between a stem cell and a terminally differentiated cell (Crosta, 2010). iv
  • 7. With the therapeutic application of NSCs for neurorestoration in mind, a clearer picture is emerging. Both in normal neurodevelopment and stem cell biology, the precursor cells display preprogrammed behavior modified by cues from the local environment. The fundamental assumption is that differentiation and predictable behavior of NSCs can be achieved if the appropriate cocktail of soluble/diffusible or contact-mediated signals is present. In addition, several corollary considerations are quickly evident. For example, can we use NSCs from different sources in an equivalent fashion? The answer to this important question requires that we understand the developmental potential of all the types of NSCs (Marquez et al., 2005). Medical researchers believe that stem cell therapy has the potential to dramatically change the treatment of human disease. A number of adult stem cell therapies already exist, particularly bone marrow transplants that are used to treat leukemia. In the future, medical researchers anticipate being able to use technologies derived from stem cell research to treat a wider variety of diseases including cancer, Parkinson's disease, spinal cord injuries, Amyotrophic lateral sclerosis, multiple sclerosis, and muscle damage, amongst a number of other impairments and conditions (Goldman and Windrem, 2006). v
  • 8. Aim of the work: The aim of this work is to study and summarize recent progress in stem cell therapies aimed at neurodegenerative disorder and illustrate how some of aforementioned methods and strategies are being utilized to formulate clinically viable treatments. vi
  • 9. Stem cells Definition: A stem cell is a cell that has the ability to divide (self replicate) for indefinite periods, often throughout the life of the organism. Under the right conditions, or given the right signals, stem cells can give rise (differentiate) to the many different cell types that make up the organism. That is, stem cells have the potential to develop into mature cells that have characteristic shapes and specialized functions, such as heart cells, skin cells, or nerve cells (Charron et al., 2009). The word “stem” actually originated from old botanical monographs from the same terminology as the stems of plants, where stem cells were demonstrated in the apical root and shoot meristems that were responsible for the regenerative competence of plants. Hence also the use of word “stem” in “meristem” (Kiessling and Anderson, 2003). Historical overview of stem cell therapy: The stem cell is the origin of life. As stated first by the great pathologist (Rudolph Virchow), “All cells come from cells”. The fertilized egg is formed from fusion of the haploid progeny of germinal stem cells. The fertilized egg is totipotent; from it forms all the tissues of the developing embryo. During development of the embryo, germinal stem cells are formed, which persist in adult to allow the cycle of life to continue. In 1
  • 10. the adult, tissue is renewed by proliferation of specialized stem cells, which divide to form one cell that remains a stem cell and another cell that begins the process of differentiation to specialized function of a mature cell type, normal tissue renewal is accomplished by the differentiating progeny of stem cells, the so-called transit amplifying cells. For example, blood cells are mature cells derived from hematopoietic stem cells in the bone marrow; the lining cells of the gastrointestinal tract are formed from transit amplifying cells, progeny of stem cell in the base of intestinal glands (Crosta, 2010). Nineteenth century pathologists first hypothesized the presence of stem cells in the adult as “embryonal rests” to explain the cellular origin of cancer and the studies indicate that the most cancers arise from stem cells or their immediate progeny, the transit-amplifying cells. Cancer results from an imbalance between the rate at which cells are produced and the rate at which they terminally differentiate or die. Understanding how to control the proliferation and differentiation of stem cells and their progeny is not only the key to controlling and treating cancer, but also to cell replacement and gene therapy for many metabolic, degenerative, and immunological diseases (Virchow, 1985). Stem cell properties: Stem cells have a capacity for self-renewal giving rise to more stem cells, and the ability to differentiate into tissues of 2
  • 11. various lineages under appropriate conditions. They may be totipotent, pluripotent or multipotent, depending on type. Only the embryo is totipotent. Embryonic stem cells (ESCs) are pluripotent, as they are capable of differentiating into many tissue types, whereas differentiation of adult stem cells is generally restricted to the tissue in which they reside, as with hepatocytes in the liver, and haemopoietic stem cells in blood (figure 1) (Bongso and Lee, 2005). Figure (1): Stem cell self-renewal and differentiation (Bongso and Lee, 2005). 3
  • 12. A) Stem cell self renewal: The defining feature of a true stem cell is the capacity for self-renewal. Self renewal occurs when a cell that has been activated to divide does so asymmetrically. The result produces one cell that is exactly like the mother cell and one cell that takes on biological functions that are different from those of the mother cell. Without self-renewal, each activation event would result in the progressive loss of the originating stem cell population (Andeson et al., 2001). B) The stem cell life cycle: Stem cell activation is generally followed by a clonal expansion of the daughter cell that is produced. This is associated with a series of biological processes that include proliferation, migration, differentiation, and at some point cell death. Regulation of these downstream events determines the net effect that, each stem cell activation has on new tissue formation (Song et al., 2007). C) Stem cell plasticity: The term plasticity means that a stem cell from one adult tissue can generate the differentiated cell types of another tissue. At this time, there is no formally accepted name for this phenomenon in the scientific literature. It is variously referred to as “plastisity” “unorthodox differentition” or “transdifferentiation” (figure 2) (Joanna et al., 2009). 4
  • 13. To show that the adult stem cells can generate other cell types requires them to be tracked in their new environment, whether it is in vitro or in vivo. In general, this has been accomplished by obtaining the stem cells from a mouse that has been genetically engineered to express a molecular tag in all its cells. It is then necessary to show that the labeled adult stem cells have adopted key structural and biochemical characteristics of the new tissue they are claimed to have generated (Gussoni et al., 2002). Also it is necessary to demonstrate that the cells can integrate into their new tissue environment, survive in the tissue, and function like the mature cells may assume the characteristic of cells that have developed from the same primary germ layer or a different germ layer, for example, much plasticity experiments involve stem cells derived from bone marrow, which is a mesodermal derivative. The bone marrow stem cells may then differentiate into another mesodermally derived tissue such as skeletal muscle, cardiac muscle or liver (Kocher et al., 2001). Stem cell lineage differentiation and commitment is conventionally viewed as progressively downstream, unidirectional and irreversible. The notion of unidirectional tissue-lineage commitment of stem cells is being challenged by evidence of plasticity, or lineage conversion, in adult stem cells. Mechanisms allowing for such plasticity include trans- differentiation which describes the conversion of a cell of one 5
  • 14. tissue lineage into a cell of an entirely distinct lineage, with concomitant loss of the tissue-specific markers and function of the original cell type, and acquisition of markers and function of the trans-differentiated cell type (Bianco et al., 2005). Alternatively, adult stem cell may differentiate into a tissue that, during normal embryonic development, would arise from a different germ layer. For example, bone marrow derived cells may differentiate into neural tissue, which is derived from embryonic ectoderm and neural stem cell lines cultured from adult brain tissue may differentiate to form hematopoietic cells, Figure (2): Evidence of plasticity of stem cell (Joanna et al., 2009). 6
  • 15. or even give rise to many different cell types in embryo. In both cases cited above, the cells would be deemed to show plasticity, but in the case of bone-marrow stem cells generating brain cells, the finding is less predictable (Song et al., 2007). Alternative mechanisms for explaining apparent stem cell plasticity involve cell-cell fusion between a stem cell and a tissue specific cell, the existence of multiple stem cell populations in one pool of cells, and the ability of the stem cells to differentiate to a more primitive, less specialized cell lineage, and then re-differentiate down another lineage (Bongso and Lee, 2005). The differentiation potential of stem cells: Many of the terms used to define stem cells depend on the behavior of the cells in the intact organism (in vivo), under specific laboratory conditions (in vitro), or after transplantation in vivo, often to a tissue that is different from the one from which the stem cells were derived (Joanna et al., 2009). So they are three classes of stem cells exist: totipotent, pluripotent multipotent and unipotent. 1) Totipotent: Totipotency is the ability of a cell to divide and produce all of the undifferentiated cells within an organism, from the Latin word totus, meaning entire; For example, the fertilized egg is said to be totipotent, because it has the potential to generate all the cells and tissues that make up an embryo and that support its development in uterus. After fertilization, the cell begins to 7
  • 16. divide and produce other totipotent cells; these totipotent cells begin to specialize within a few days after fertilization. The totipotent cells specialize into pluripotent cells, which they develop into the tissues of the developing body. Pluripotent cells can further divide and specialize into multipotent cells, which produce cells of a particular function (Svendsen and Ebert, 2008). Adult mammals, including humans, consist of more than 200 kinds of cells. These include nerve cells (neurons), muscle cells (myocytes), skin (epithelial) cells, blood cells (erythrocytes, monocytes, lymphocytes, etc.), bone cells (osteocytes) and cartilage cells (chondrocytes). Other cells, which are essential for embryonic development but are not incorporated into the body of the embryo, include the extraembryonic tissues, placenta, and umbilical cord. All of these cells are generated from a single, totipotent cell, the zygote or fertilized egg (Joanna et al., 2009). 2) Pluripotent: Pluripotent stem cells can give rise to any type of cell in the body except those needed to develop a fetus or adult because they lack the potential to support the extraembryonic tissue (e.g., the placenta). Most scientist use the term pluripotent to describe stem cells that can give rise to cells derived from all three embryonic germ layers (endoderm, mesoderm, and 8
  • 17. ectoderm). These three germ layers are the embryonic source of all cells of the body (figure 3) (Svendsen and Ebert, 2008). Figure (3): Pluripotent stem cells (Svendsen and Ebert, 2008). The term “pluri” is derived from the Latin word plures, means several or many. Thus, pluripotent cells have the potential to give rise to any type of cell, a property observed in the natural course of embryonic development and under certain laboratory conditions. Pluripotent stem cells are isolated from embryos that are only several days old; cells from these stem 9
  • 18. cell lines can be cultured in the lab and grown without limit (Sonja et al., 2006). 3) Multipotent: Multipotent cells, in contrast, can only give rise to a small number of cell types and they can produce only cells of a closely related family cell. As haematopiotic stem cells that differentiate to red blood cells, white blood cells and platelets. A hematopoietic cell, or a blood stem cell, can develop into several types of blood cells but cannot develop into liver cells or other types of cells; the differentiation of the cell is limited in scope. A multipotent blood cell can produce red and white blood cells (figure 4) (Svendsen and Ebert, 2008). Figure (4): Multipotent stem cell (Svendsen and Ebert, 2008). 10
  • 19. 4) Unipotent: Unipotent stem cells, a term that is usually applied to a cell in adult organisms, means that the cells in question are capable of differentiating along only one lineage. The term “uni” is derived from the Latin word unus, which means one. Also, it may be that the adult stem cells in many differentiated, undamaged tissues are typically unipotent and give rise to just one cell type under normal conditions. This process would allow for a steady state of self renewal for the tissue. However, if the tissue becomes damaged and the replacement of multiple cell types is required, pluripotent stem cells may become activated to repair the damage (Avasthe et al., 2008). F igu re (5): Differentiation of human stem cells (Bongso and Lee, 2005). 11
  • 20. Classification of stem cells according to their sources: Stem cells can be classified into four broad types based on their origin, stem cells from embryos; stem cells from the fetus; stem cells from umbilical cord; and stem cells from the adult. Each of these can be grouped into subtypes (Andeson et al., 2001). 1) Embryonic stem cells: In mammals; the fertilized oocyte, zygote, 2-cells, 4-cells, 8- cells and morula resulting from cleavage of the early embryo are examples of totipotent cells (ability to form a complete organism) (figure 6) (Avasthe et al., 2008). 12
  • 21. Figure (6): Development and differentiation of human tissues (Avasthe et al., 2008). The inner cell mass (ICM) of the 5 to 6 days old human blastocyte is the source of pluripotent embryonic stem cells (HESCs) and consisting of 50–150 cells (figure 7) (Bongso and Lee, 2005). 13
  • 22. Figure (7): Human blastocyst showing inner cell mass and trophectoderm (Bongso and Lee, 2005). 14
  • 23. Figure (8): How human embryonic stem cells are derived? (Bongso and Lee, 2005). Characteristics of human embryonic stem cells: They can maintain undifferentiated phenotype and these cells are able to renew themselves continuously through many passages leading to the claim that they are immortal, also these 15
  • 24. cells are pluripotent, meaning that they are able to create all three germ layers of the developing embryo and thus they can develop into each of the more than 200 cell types of the adult body (figure 9) (Junying et al., 2006). Figure (9): Characteristics of embryonic stem cells (Junying et al., 2006). 16
  • 25. Nearly all research to date has taken place using mouse embryonic stem cells (MES) or Human embryonic stem cells (HESCs). Both have the essential stem cell characteristics, yet they require very different environments in order to maintain an undifferentiated state. Mouse ES cells are grown on a layer of gelatin and require the presence of Leukemia Inhibitory Factor (LIF) (Bongso and Lee, 2005). HESCs are grown on a feeder layer of mouse embryonic fibroblasts (MEFs) and require the presence of basic Fibroblast Growth Factor (bFGF or FGF-2). Without optimal culture conditions or genetic manipulation, embryonic stem cells will rapidly differentiate (Avasthe et al., 2008). Identification of the human embryonic stem cells: Laboratories that grow human embryonic stem cell lines use several kinds of tests to identify the human embryonic stem cells. These tests include: 1- Growing and sub-culturing the stem cells for many months. This ensures that the cells are capable of long term self-renewal. Scientists inspect the cultures through a microscope to see that the cells look healthy and remain undifferentiated (Lawrence et al., 2006). 2- Using specific techniques to determine the presence of surface markers that are found only on undifferentiated cells. Another important test is for the presence of a 17
  • 26. protein called oct-4, which undifferentiated cells typically make. Oct-4 is a transcription factor, meaning that it helps turn genes on and off at the right time, which is an important part of the processes of cell differentiation and embryonic development. 3- Examining the chromosomes under a microscope. This is a method to assess whether the chromosomes are damaged or if the number of chromosomes has changed. It does not detect genetic mutations in the cells. 4- Determining whether the cells can be subculture after freezing, thawing, replanting (junying et al., 2006). Differentiation of human embryonic stem cells: In order to start differentiation, the HESCs must be removed from the feeder layer and the cell replated and will form embryoid bodies (Ebs), spherical aggregates in which the HESCs undergo mixed spontaneous differentiation toward lineages of all three dermal layers. Another protocol of differentiation directly without formation of embryoid bodies stage have resulted in more controlled differentiation and better yield of the required cells (figure 10) (Joanna et al., 2009). 18
  • 27. Figure (10): Fluorescent markers can be used to identify stem cells hidden among ordinary adult cells. Here, human embryonic stem cells are recognized by the marker proteins they express (green) (Joanna et al., 2009). Ethical considerations: The promise of stem cell therapy has ignited public dispute on the ethics of using aborted embryos for medical purposes. Individual attitudes are usually influenced by religious and liberal views but also by concerns that the practice of embryonic tissue transplantation will increase the pressure to perform abortions and create a black market in which pregnancy and aborted tissues will be sold to the highest bidder. The regulated banking of stem cell lines may solve some of the ethical issues. As in other cases in which medical and scientific advances found society without the means to deal with their ethical, legal, and social consequences, it is important to discuss these issues in public, with the active participation of the medical and scientific community (Christopher, 2008). 19
  • 28. 2) Fetal stem cells: The identification of human fetal stem cells has raised the possibility of using autologus cells for in utero treatments. The human fetal stem cells population extracted from fetal blood contains adherent cells that divide in culture for 20 to 40 passages and can differentiate into mesenchymal lineages including bone and cartilage, but also have the ability to form oligodendrocytes and hematopoiotic cells. These cells, which can be found circulating only during the first trimester, are similar to hematopoiotic populations in fetal liver and bone marrow (Avasthe et al., 2008). 3) Umbilical cord stem cells: These are cells harvested from the cord blood. Cord blood is rich in the stem cells and after appropriate human leukocyte antigen [HLA] matching may be used to treat a variety of conditions. Characteristics of these cells are identical to adult stem cells except that they are not derived from adults and that their concentration is far more in umbilical blood as compared to adults. The use of umbilical cord stem cells in orthopedics is still in a nascent stage and most studies currently focus on the use of the stem cell (Crosta, 2010). 4) Adult stem cell: It is an undifferentiated cell that is found in a differentiated tissue, it can renew itself and become specialized to yield all the 20
  • 29. specialized cell types of the tissue from which it originated. Adult stem cells, like all stem cells, share at least two characteristics. First, they can make identical copies of themselves for long period of time; this ability to proliferate is referred to as long term self renewal. Second, they can give rise to mature cell types that have characteristic morphologies (shapes) and specialized functions (Charron et al., 2009). Typically, stem cells generate an intermediate cell type or types before they achieve their fully differentiated state. The intermediate cell is called a precursor cells in fetal or adult tissues are partially differentiated cells that divide and give rise to differentiated cells. Such cells are usually regarded as “committed” to differentiating along a particular cellular development pathway, although this characteristic may not be as definitive as once thought (Bianco et al., 2005). Adult stem cells are rare. Their primary functions are to maintain the steady state functioning of a cell, called (homeostasis) and with limitation to replace cells that die due to injury or disease. For example, only an estimated 1 in 10,000 to 15,000 cells in the bone marrow is a hematopoietic (blood- forming) stem cell (HSC). Furthermore, adult stem cells are dispersed in tissues throughout the nature of animal and behave very differently, depending on their local environment. For example, HSCs are constantly being generated in the bone marrow where they differentiate into mature types of blood 21
  • 30. cells. Indeed, the primary role of HSCs is to replace blood cells (Abdallah and Kassem 2008). Unlike embryonic stem cells, which are defined by their origin (the inner cell mass of the blastocyte), adult stem cells share no such definitive means of characterization. In fact, no one knows the origin of adult stem cells in any mature tissue. Some have proposed that stem cells are somehow set aside during fetal development and restrained from differentiating. Definition of adult stem cells vary in the scientific literature range from a simple description of the cells to a rigorous set of experimental criteria that must be met before characterizing a particular cell as an adult stem cell. Most of the information about adult stem cells comes from studies of mice. The list of adult tissues reported to contain stem cells is growing and includes bone marrow, peripheral blood, brain, spinal cord, dental pulp, blood vessels, skeletal muscle, epithelia of skin and digestive system, cornea, retina, liver, and pancreas (Christopher, 2008). Ideally, adult stem cells should also be clonogenic. In other words, a single adult stem cell should be able to generate a line of genetically identical cells, which then gives rise to all the appropriate, differentiated cell types of the tissue in which it resides. Again, this property is difficult to demonstrate in vivo; in practice, scientists show either that a stem cell is clonogenic in vitro, or that a purified population of candidate stem cells can repopulate the tissue (Avasthe et al., 2008). 22
  • 31. Sources of adult stem cells: (I) Bone Marrow-Derived Stem/Progenitor Cells: Adult bone marrow-derived stem cells are presently the cell types most widely used in stem cell therapy. A heterogeneous subset there of, termed autologous bone marrow-derived mononuclear cells (ABMMNCs), comprises the following types of stem cells, (Mesenchymal stem cells, Hematopoietic stem cells and Endothelial progenitor cells), that have potential therapeutic uses (figure 11) (Svendsen and Ebert, 2008). Figure (11): Some of the known sources of adult stem cells (Svendsen and Ebert, 2008). 23
  • 32. (a) Mesenchymal stem cells (MSCs): MSCs are a proper stem cell which can be greatly and efficiently expanded in culture and can differentiate to several specific mesenchymal cell lineages. Mesenchymal (Stromal) stem cells (MSCs) are found in various niches of adult tissue. MSCs are rare in bone marrow (<0.01% of nucleated cells, by some estimates) and 10 times less abundant than hematopoietic progenitor cells but MSCs can be readily grown in culture. However, more recently, other sources of MSCs have been described including placenta, adipose tissue, cord blood and liver (junying et al., 2006). The human Mesenchymal stem cells (HMSCs) from bone marrow can be cloned and expanded in vitro more than 1 million-fold and retain the ability to differentiate to several mesenchymal lineages. Researchers have not yet found conditions that allow continuous, indefinite HMSC growth, yet it is possible to produce billions of MSCs in vitro for cellular therapy from a modest bone marrow aspirate drawn through the skin. MSCs need to be expanded ex vivo because they apparently are very contact inhibited, and there is little evidence of in vivo expansion as MSCs labeled with membrane dyes, that would be diluted and undetected from dividing cells after about 3 divisions, are found months later even in repairing tissue (Sottile et al., 2002). 24
  • 33. Advantages of Mesenchymal Stem Cells: Ease of isolation, high expansion potential, genetic stability, reproducible characteristics in widely dispersed laboratories, compatibility with tissue engineering principles and potential to enhance repair in many vital tissues. There they may be the current preferred stem cells model for cellular therapeutic development (Diana and Gabriel, 2008). Biology of mesenchymal stem cells (MSCs): The anatomical locations of phenotype of MSCs have no yet been well defined in vivo. Some have used expression of Stro-1 and VCAM-1 to analyses putative MSC in vivo in human. A general consensus among researchers in the field is that MSC can be successfully defined based on staining with surface markers such as CD44, CD90, CD73, CD105 and CD166. However, none of these antigens are unique to MSC. Using markers such as Stro-1 (human) and Sca-1 (mouse), several reports indicate that MSC reside adjacent to endothelium in the bone marrow and possibly other tissues (Zannettino et al., 2007). (b) Hematopoietic stem cells (HSCs): HSCs are presented in umbilical cord blood with a frequency of just under one in 1 million mononuclear cells (one in 3 million MNCs) or mobilized peripheral blood (one in 6 million MNCs). They are capable of unlimited cell proliferation in bone marrow and must undergo at least 20 to 23 divisions on their 25
  • 34. way to produce mature blood cells, even assuming no cell death along the way (Emerson et al., 2008). Biology of heamtopiotic stem cells: Much effort has been focused on discovering cell surface markers that can identify those cells that have true functional stem cell properties. Perhaps clinically most familiar is CD34, a glycoprotein present on the cell surface of stem and progenitor cells which is used to enrich stem cells mobilization and collection for HSCs, but even within the CD34+ population, only a small percentage are HSCs (Emerson et al., 2008). For decade scientists and hematologists have struggled with the difficulty that HSCs cannot be purified based on phenotypical characteristics and perhaps more importantly, cannot be expanded and cloned ex vivo. Recent evidence has emerged suggesting that HSCs can be expanded ex vivo. But there is still no evidence to support the idea of clonality. For these reasons HSCs are not ideally suited for in vitro experiments designed to test plasticity. In this regard HSCs differ dramatically from MSCs in bone marrow and neural stem cells (NSCs) in the central nervous system, both of which can be clonally derived and tested for multiple differentiation pathways (figure 12) (Joanna et., 2009). 26
  • 35. Figure (12): Hematopoietic and stromal stem cell differentiation (Joanna et al., 2009). (c) Endothelial progenitor cells (EPCs): A subset of bone marrow-derived hematopoietic progenitor cells: endothelial progenitor cells (EPCs). These cells can give rise to endothelial recovery and new capillary formation after ischemia (Einstein and Ben-Hur, 2008). (II) Neural stem cells: The concept that adult mammalian CNS contains NSCs was first inferred from evidence of neuronal turnover in olfactory 27
  • 36. bulb and hippocampus in the adult organism. The multipotency of NSCs was demonstrated in vitro in 1990 by their ability to differentiate into neurons, astrocytes, and oligodendrocytes as well as various forms of neural precursors. In addition, in vivo delivery of these cells to animal models of neurodegenerative diseases was associated with varying degrees of functional recovery. Currently, there is no set of markers or protein expression profiles that precisely define and fully characterize undifferentiated NSCs. Neural stem cells (NSCs) and neural precursor cells (NPCs) can be isolated from the developing or adult CNS and can be safely expanded in chemically defined culture media for an extended (Song et al., 2007). (a) Adult neural precursor cells (NPCs): New neurons are derived in adulthood from a population of adult NPCs, which are primarily found in the subependymal layer of the ventricular zone and the dentate gyrus of the hippocampus, although they are also probably found in other sites. However, the behavior of the neural precursor cells (NPCs) found in all these sites is different, and may relate as much to the environment in which they find themselves as to their intrinsic properties, eg; nigral NPCs appear to only differentiate into astrocytes in situ or when grafted to the adult nigra, but when they are cultured in vitro or transplanted into the hippocampus they can form neurons (Gronthos et al., 2003). 28
  • 37. Properties of neural stem cells: (1) Immunosupressive effect of NSCs: Although NSCs may exert their therapeutic effects by directly replacing missing cells, transplantation rarely results in significant numbers of transplant-derived terminally differentiated neurons. The beneficial effect of NSCs in disease models may be attributable to alternative biologic properties. The first indication of an anti-inflammatory effect of NPCs came from transplantation experiments in rats with experimental autoimmune encephalomyelitis (EAE). It was shown transplantation of NPCs reduced brain inflammation and clinical disease severity, it was suggested that the benefit of NPC transplantation was mediated by an anti-inflammatory effect (Raisman and Li, 2007). The exact mechanisms by which transplanted NPCs attenuate brain inflammation are unclear. Some suggests an immunomodulatory effect by which NPCs promote apoptosis of type 1 T-helper cells, shifting the inflammatory process in the brain toward a more favorable climate of dominant type 2 T- helper cells. Alternatively, a nonspecific bystander immunosuppressive effect of NPCs on T-cell activation and proliferation has been suggested. The suppressive effect of NPCs on T cells was accompanied by a significant suppression of pro-inflammatory cytokines. This nonspecific anti- inflammatory mechanism may be of major importance in the application of transplantation therapy in immune-mediated 29
  • 38. diseases because it can protect the host CNS and graft from additional immune attacks (Einstein and Ben-Hur, 2008). (2) Neuroprotictive effects of transplanted NSCs: Neuroprotective effect was observed in other non autoimmune experimental disease models. Neural stem cells rescued dopaminergic neurons of the mesostriatal system in a Parkinson disease (PD) model in rodents. These findings led to the concept that NSCs are endowed with inherent mechanisms for rescuing dysfunctional neurons. This effect was found to be important in other neurologic diseases. Neural stem cells seeded on a synthetic biodegradable scaffold and grafted into the hemi- sectioned adult rat spinal cord induced significant improvement in animal movement by reduction of necrosis in the surrounding parenchyma and by prevention of inflammation, glial scar formation, and extensive secondary cell loss (Einstein and Ben- Hur, 2008). (3) Neurotrophic effects of transplanted NSCs: After sectioning of the adult spinal cord, NSC transplantation induced a permissive environment for axonal regeneration. Similarly, in a model of retinal degeneration, NPC transplantation promoted neural growth in the optic nerve. In both cases, this effect was mediated by induction of matrix metalloproteinases that degrade the impeding extracellular matrix and cell surface molecules, enabling axons to extend through the glial scar. Transplantation of olfactory-ensheathing cells into the sectioned spinal cord also promoted axonal 30
  • 39. regeneration in long fiber tracts, with a return of lost function. This was explained by the creation of proper realignment, enabling axonal growth through a permissive tract. In addition, the cells increased axonal sprouting, remyelination, and vascularization of the injured spinal cord (Raisman and Li, 2007). Isolation of human NSCs: To date, they are primary isolated and propagated in vitro as cells that form free-floating neurospheres when cultured in serum-free medium on non adherent surfaces in the presence of mitogenic factors such as basic FGF or FGF-2 and epidermal growth factors, although there have also been reports of monolayer cultures (McBride et al., 2004). (III) Pancreatic stem/progenitor cells: There is strong evidence that new pancreatic islets can derive from progenitor cells present within the ducts and islets, in a process called “neogenesis”. Furthermore, when these pseudo-islets were transplanted into non-obese diabetic (NOD) mice, diabetes reversal was observed. Candidate pancreatic stem/progenitor cells have also been described within acini, but contamination with endocrine and ductal cells in cultures could not be excluded in these experiments (Limmbert et al., 2008). The isolation of a distinct stem/progenitor cell within the endocrine pancreas depends on the identification of a specific progenitor marker. The exciting observation that nestin positive islets cells display endocrine differentiating capacity led to the 31
  • 40. hypothesis that this intracytoplasmic filament protein might correspond to a pancreatic stem/progenitor cell marker. More recently, in two important studies a population of cells in the developing and adult mouse pancreas was identified, which under differentiation conditions, released insulin in a glucose- dependant manner. After differentiation, these cells expressed specific developmental pancreatic endocrine genes (e.g. Ngn3, Pax-4, Pax-6 and PDX-1) and contamination with mature beta cells was ruled out (Limbert et al., 2008). While mature beta cell replication appears to be major physiological beta-cell regenerative process, identification of pancreatic cells with progenitor features might open an important and promising strategy for cell replacement and regeneration therapy. Anyhow, to be clinically relevant, in vitro proliferation of progenitor cells from human pancreas must produce large amounts of cells, in order to allow cells isolated from one single donor to be sufficient to treat a given diabetic patient. It would be even better to have one single donor for several diabetics. For these reasons, acinar isolated stem/progenitor cells might be of interest, considering that exocrine tissue constitutes 90% of pancreatic tissue and is discarded during islet isolation (Kushner et al., 2005). (IV) Other sites: First identified in human bone marrow, a population of mesenchymal progenitor/stem cells (MSC) with well characterized immunophenotype and distinct from 32
  • 41. hematopoietic stem cells, was shown to possess a high proliferation rate and great plasticity. Under specific culture conditions these cells differentiate into mesenchymal tissues, such as bone, cartilage, muscle, tendon, adipose and stroma, as well as neuronectodermal tissues (Limbert et al., 2008). Adult tissues and organs known to have stem cells Source Description Brian Stem cells of the brain can differentiate into the three kinds of nervous tissue-astrocytes, oligodendrocytes, and neurons-and in some cases, blood-cell precursor. Bone marrow These occur as hematopoietic stem cells, which give rise to all blood cells, and as stroma cells, which differentiate into cartilage and bone. Endothelium These stem cells are called hemangioblasts and are known to differentiate into blood vessels and cardiomyocytes. They may originate in bone marrow, but this is uncertain. Skeletal muscle These stem cells may be isolated from muscle or bone marrow. They mediate muscle growth and may proliferate in response to injury or exercise. Skin Stem cells of the skin are associated with the epithelial cells, hair follicle cells, and the basal layer of the epidermis. These stem cells are involved in repair and replacement of all types of skin cells. Digestive Located in intestinal crypts, or invaginations. These stem cells are system responsible for renewing the epithelial lining of the gut. Many types are believed to exist, but examples have yet to be Pancreas isolated. Some neural cells are known to generate pancreatic β cells. The identity of liver stem cells is still unclear. Stem cells from bone Liver marrow may repair some liver damage, but most repairs seems to be carried out by the hepatocytes (liver cells) themselves. Table (1): Sources of adult stem cells (Limbert et al., 2008). 33
  • 42. Identification of the adult stem cells: The scientists often use one or more of the following three methods to identify and test adult stem cells: 1- Labeling the cells in a living tissue with molecular markers and then determining the specialized cell types they generate. Then 2- Removing the cells from living animals, labeling them in cell culture, and transplanting them back into another animal to determine whether the cells repopulate their tissue of origin. Then 3- Isolating the cells, growing them in cell culture, and manipulating them, often by adding growth factors or introducing new genes, to determine what differentiated cells types they can become (Raisman and Li, 2007). The similarities and differences between embryonic and adult stem cells: The adult and embryonic stem cells differ in the number and types of differentiated cells types they can become. Embryonic stem cells can become all cell types of the body because are pluripotent. Adult stem cells are generally limited to differentiating into different cell types of their tissue of origin. However, some evidence suggests that adult stem cell plasticity may exist; increasing the number of cell types a given adult stem cell can become (figure 13). Large numbers of embryonic stem cells can be relatively easily grown in culture, while adult stem cells are rare in mature tissues and methods for expanding 34
  • 43. their numbers in cell culture have not yet been worked out. This is an important distinction, as a large number of cells are needed for stem cell replacement therapies (Limbert et al., 2008). A potential advantage of using stem cells from an adult is that the patient’s own cells could be expanded in culture and then reintroduced into the patient. The use of patient’s own adult stem cells would means that the cell would not be rejected by the immune system. This represents a significant advantage as immune rejection is a difficult problem that can only be circumvented with immunosuppressive drugs. Embryonic stem cells from a donor introduced into a patient could cause transplant rejection, however, whether the recipient would reject donor embryonic stem cells has not been determined in human experiments (Sonja et al., 2006). Figure (13): Sources of stem cells (Limbert et al., 2008). 35
  • 44. Types of Stem Cell transplantation: Stem cell transplantation can be classified according to the genetic relation between the donor and recipient into 4 classes: 1- Autograft: In which the donor and recipient is the same individual. 2- Isograft or syngenic graft: In which the donor and recipient are genetically identical (e.g., monozygotic twins). 3- Allograft or homograft: In which the donor and recipient are genetically unrelated but belong to the same species. 4- Xenograft or heterograft: In which the donor and recipient belong to different species (David, 2009). Application of stem cells: 1) Basic science application: Stem cells are ideally suited to allow for the study of complex processes that direct early unspecialized cells to differentiate and develop into the more than two hundred cell types in the human body (Bianco et al., 2005). 2) Medical research applications: Stem cell studies may allow researchers to follow the processes by which diseases and impairments caused by genetic abnormalities first manifest themselves biochemical or structurally in cells and tissues. Using stem cells to produce large numbers of genetically uniform cultures of organ tissues for example, liver, muscle, or neural would allow controlled comparison of the effects of drugs or chemical on these tissues. 36
  • 45. Alternatively, testing drugs against stem cell tissues varying genetic makeup could allow tissue specific stem cell may provide a constant in vitro source of such cellular material (Bianco et al., 2005). The site of stem cell implantation: The transplantation can be described as orthotropic or heterotropic: 1- Neurologic transplantation: Refers to donor tissue implantation in the anatomically correct position in the recipient. 2- Heterotropic transplantation: Refers to the relocation of the implant in the recipient at a site different from the normal anatomy (David, 2009). Route of stem cell delivery: Reports have indicated that after stereotactic intraparenchymal, intracerebro-ventricular, intravenous and intraarterial transplantation, stem cells can home to sites of injury in the CNS and induce functional recovery. Of these various transplantation techniques, those that depend on intravascular delivery of stem cells for stroke are particularly attractive. Intravascular delivery: In addition to its minimal invasive nature, intravascular delivery may allow stem cells to have a superior interaction with injured tissue. A comparative study revealed that direct intracerebral transplantations resulted in the largest number of 37
  • 46. cells at the lesion site, followed by intracerebro-ventricular and intravenous transplantations (Guzman et al., 2008). However, researchers in that study only assessed the absolute number of cells in the perilesional area and took no account of whether these cells were therapeutically distributing to all injured areas of brain parenchyma on a microscopic level. Many believe that intravascular delivery of stem cells may lead to a wider distribution of cells around the lesion as compared with focal perilesional transplants, thereby leading to superior stem cell–injured tissue interactions (Xiao et al., 2007). Mechanism of wide distribution: The cells travel in the blood stream and follow a chemo attractant gradient generated by inflammation in the injured brain. Unfortunately, intravenously delivered cells pass through the systemic and pulmonary circulation systems and home to other organs as well, which significantly reduces cell homing to the injured brain. Intravenous injection of human MSCs into rats 24 hours after stroke showed that only 4% of the cells entered the brain, the number of cells entering the brain increased over time and peaked at Day 21 post-stroke. At Day 56, 60% of these surviving cells differentiated into glia, and 20% into neurons. Despite the fact that the number of cells entering the brain was limited, functional recovery was enhanced by intravenous delivery (Pluchino et al., 2005). 38
  • 47. Intracarotid injection: Another route of intravascular delivery is intra arterial, which would circumvent body circulation. The first pass of stem cells injected into the carotid artery would be the brain, this route of delivery have demonstrated functional recovery after stroke and traumatic brain injury. In 2006 Shen and colleagues injected donor rat BMSCs into the internal carotid artery of rats 24 hours post-stroke and successfully induced functional recovery. In another study, the same group injected donor rat BMSCs into rats 24 hours after stroke and observed that injected cells localized around the infarction area in the brain and very few were found in the heart, lungs, liver, spleen, and kidney (figure 14) ( Guzman et al., 2008). Figure (14): Confocal laser scanning microscopy images revealing numerous cells in the stroke border zone and the hippocampus ipsilateral to the stroke (A). Inset shows doublecortin bromodeoxyuridine labeled cells. The VCAM-1 (arrows) is highly expressed in the stroke affected hemisphere 48 hours after stroke (B). DCX = doublecortin; BrdU = bromodeoxyuridine; DAPI = 4'6-diamidino-2-phenylindole (Guzman et al., 2008). 39
  • 48. The debate over the best delivery route is further complicated by the fact that there is still a great deal of controversy concerning the mechanism by which stem cells lead to enhanced functional recovery in patients who have experienced stroke. The 2 most discussed mechanisms are as follows: 1) cellular replacement, by way of the functional integration of stem cells; and 2) secretion of neurotrophic and angiogenic factors. If the mechanism of recovery is cellular replacement, then transendothelial migration is necessary and the methods that allow the highest concentrations of stem cells in the injured brain areas ought to be pursued; however, there is significant evidence that stem cells may provide their benefits by secreting various neuroprotective factors (Guzman et al., 2008). In summary, the best route of human stem cell delivery has not been determined, but the intravascular route is particularly attractive because of its ease of administration, minimal invasiveness, and potential for widespread cell distribution together with widespread secretion of neuroprotective, proangiogenic, and immunomodulatory factors. Intuitively, the intraarterial route of delivery seems better than the intravenous, given that injected cells first pass the target organ that is, the brain prior to being redistributed in the systemic circulation (Pluchino et al., 2005). 40
  • 49. Timing of transplantation Undoubtedly the fate and function of transplanted cells will depend on any or all of these alterations, and the optimal time of transplantation is unknown. The timing of transplantation depends mainly on the goal of treatment, for example, neuroprotection, which should happen early after the insult, or neuroregeneration/cell replacement, which can be done once a lesion has stabilized. We can envision a future in which we will rely on multimodal stem cell treatment, depending on a combination of early and late administrations of different cell types (Guzman et al., 2008). Early intravascular cell delivery In animal models with a neuro inflammatory component such as stroke, traumatic brain injury, spinal cord injury, and multiple sclerosis, therapeutic somatic stem cells (for example, BMSCs, umbilical cord blood stem cells, MSCs, and NPCs) target inflamed CNS areas where they persist for months and promote recovery through neuroprotective mechanisms. It is thought that the process of transendothelial migration of somatic stem cells may be regulated in a manner similar to that of inflammatory cells. As early as 30 minutes after stroke, the infiltration of leukocytes, both polymorphonuclear leukocytes and monocytes/macrophages, can be observed (Goldman and Windrem, 2006). Chemoattraction, adhesion, and transendothelial migration of inflammatory cells is regulated by specific inflammatory 41
  • 50. mediators, which have been identified in experimental and human stroke. The temporal expression profile of adhesion molecules, cytokines, and chemokines after stroke has been well described. Vascular cell adhesion molecule–1 (VCAM-1) has been shown to reach a peak level 24 hours after experimental stroke. At the bedside, soluble VCAM-1 concentration in plasma is increased in patients with acute stroke. Intercellular adhesion molecule–1 levels have been elevated as early as 4 hours after stroke with sustained levels for up to 1 week (Zhang and Lodish, 2005). Monocyte chemo-attractant protein–1, a key chemoattractant factor for the recruitment of circulating peripheral cells to the stroke area and an important factor for stem cell migration, is upregulated 3 days after stroke and then returns to baseline after 1 week. Similarly, stromal-derived factor–1(SDF-1) is known to be a potent chemoattractant for inflammatory as well as stem cells (including BMSCs and NSCs) and is expressed early after stroke. Anatomically, adhesion molecule upregulation as well as chemokine expression has been shown to be highest in the stroke-affected penumbral region. Blocking the different pathways of chemoattraction and cell adhesion in stroke- affected rodents reduced the number of infiltrating inflammatory cells (Belmadani et al., 2006). In mice lacking intercellular adhesion molecule–1 (ICAM- 1), a significant reduction in inflammatory cellular infiltrate and a reduction in lesion size were noted. Treatment with anti– 42
  • 51. ICAM-1 antibodies was a successful neuroprotective means of reducing lesion size and apoptosis in experimental stroke. However, a clinical trial exploring the feasibility of using an ICAM-1 blocking antibody failed to demonstrate any beneficial effects in the patients. There is some evidence that intravascularly administered stem cells undergo the same process as inflammatory cells, including chemoattraction, adhesion, and transendo-thelial migration after stroke, potentially making this route an ideal way of cell delivery (Hill et al., 2004). Late intraparenchymal cell transplantation In contrast to the acute intravascular cell treatment, the intraparenchymal approach has been hindered by poor outcomes if the stem cells are transplanted too early after stroke. Excitotoxicity, oxidative stress, and inflammation post ischemia make the ischemic brain a hostile environment for intracerebrally transplanted cells. In fact, we have found a negative correlation between graft survival and inflammation. Additionally, we demonstrated that human NSCs transplanted too close or into the stroke area have very limited survival at days after stroke (Belmadani et al., 2006). Transplanting cells 3 weeks after stroke, when there is a significant decrease in inflammation, led to greater graft survival than transplanting 5–7 days after stroke. Taken together, early intravascular cell therapy might benefit from the processes tied to post-stroke inflammation but might be 43
  • 52. detrimental to cells directly transplanted intra-parenchymally. Therefore, intra-parenchymal cell replacement therapy might be useful as a second line or delayed stem cell treatment strategy (Grabowski, 2010). 44
  • 53. Stem cell therapy in Parkinsonism Parkinson’s disease (PD) otherwise known as ‘’paralysis agitans’’ or ‘’shaking palsy’’ was classically described by James Parkinson in 1817. His description of “Involuntary tremulous motion with lessened muscular power, in parts not in action and even when supported, with a propensity to bend the trunk forward and to pass from a walking to a running pace, the senses and intellect being uninjured” has stood the test of time. PD is also defined as a debilitating neurodegenerative disorder of insidious onset in middle or late age characterized by the selective loss of nigrostriatal dopaminergic neurons and loss of dopamine in the striatum (Abayomi, 2002). Parkinson’s disease is second only to Alzheimer’s disease with a prevalence of 1 in 10,000. Although it is uncommon in people under age 40 years, the incidence of PD greatly increases with age, affecting approximately 1% of individuals older than 60 years (Lane et al., 2008). Pathology: The basic pathology is cell degeneration and loss of pigmented neurons in the pars compacta of the substantia nigra and locus ceruleus with atrophy and glial scarring. The degenerated pigmented neurons contain Lewy bodies which are intracytoplasmic eosinophilic hyaline inclusions composed of protein filaments (ubiquitin & synuclein), and do not have the electronic microscropic appearance of any known viral or 45
  • 54. infective agent (fig. 15). Lewy bodies are characteristic of Parkinson’s disease except in post-encephalitic Parkinson’s and parkengene mutants. However, they could be found in 4% of brain without parkinsonian features and these are likely cases of subclinical Parkinson’s as 80% of the zona compacta cells must degenerate before clinical symptoms become apparent (Abayomi, 2002). The pars compacta contains 450,000 dopaminergic neurons. With the loss of dopaminergic neurons at those sites, there is deficiency of dopamine in the basal ganglia, chiefly the striatum (caudate nucleus and putamen). Furthermore, the enzymes required for dopamine synthesis, DOPA decarboxylase and the rate limiting enzyme tyrosine hydroxylase are reduced. In addition, there is deficiency of neurotropic factors such as glial and brain derived neurotrophic factors. However, neurons in the striatum with dopamine receptors remain intact and are responsible for the therapeutic effects of levodopa. In the Parkinsonism unresponsive to levodopa, striatal neurons are degenerated. Genetic and environmental factors are important in the mechanism of neuronal deaths due to neuronal necrosis or apoptosis. In neuronal necrosis there is disintegration of cell and organelles and subsequent removal by phagocytic and inflammatory response with increased cellular permeability. In apoptosis on the other hand, there is rapid programmed cell death in response to a toxic stimuli. There is chromatin condensation, DNA fragmentation and cell shrinkage, with 46
  • 55. relative sparing of organelles without inflammatory changes or increased cellular permeability (Golbe, 2003). Among the factors that have been implicated in neuronal degeneration in Parkinson’s disease are mitochondrial dysfunction, oxidative stress, and the actions of excitotoxins, deficient neurotrophic support and immune mechanisms. HLA- DR positive reactive microglial cells and cytokines such as interleukin 1 (IL-1) and tumor necrosis factor-a play significant role in the pathogenesis of Parkinson’s disease. Oxidative stress with excess reactive oxygen species and free radical damage involving one or more unpaired electrons react with nucleic acids, proteins and lipids, this metabolic derangement results in generation of toxic byproducts and increased oxidative stress with resultant cellular damage (figure 15) (Lane et al., 2008). 47
  • 56. Figure (15): Neuronal Pathways that degenerate in Parkinson's disease. Signals that control body movements travel along neurons that project from the substantia nigra to the caudate nucleus and putamen (collectively called the striatum) (Lane et al., 2008). Cell replacement therapy: The idea of growing dopamine cells in the laboratory to treat Parkinson’s is the most recent step in the long history of cell or tissue transplantation to reverse this devastating disease. The concept was, and still is, straightforward: implant cells into the brain that can replace the lost dopamine releasing neurons. Although conceptually straight forward, this is not an easy task. Fully developed and differentiated dopamine neurons do not survive transplantation, so direct transplantation of fully developed brain tissue from cadavers, for example, is not an 48
  • 57. option. Moreover, full functional recovery depends on more than cell survival and dopamine release; transplanted cells must also make appropriate connections with their normal target neurons in the striatum (Lindvall and Kokaia, 2010). Under the basic principle of restoring dopamine producing neurons via neural grafts, extensive studies have been done to bring this to fruition. One of the first attempts at using cell transplantation in humans was tried in the 1980s. This surgical approach involved the transplantation of dopamine producing cells found in the adrenal glands, which sit atop the kidneys in the abdomen dramatic improvement in Parkinson’s patients by transplanting dopamine producing chromaffin cells from several patients’ own adrenal glands to the nigrostriatal area of their brains; it showed dramatic improvement in Parkinson’s patients. Another strategy was previously attempted in the 1970s, in which cells derived from fetal tissue from the mouse substantia nigra was transplanted into the adult rat eye and found to develop into mature dopamine neurons (Panchision, 2006). The functional integration of dopamine neuron grafts prove the efficacy of the cell replacement principle, but in reality, this clinical outcome is extremely inconsistent with respect to the percentage of cells that survive the grafting procedure and the amount of dopamine produced by the new neurons. In fact, average functional improvement of patients in the experiments only rises about 20%. Across the board, subjects achieve functioning levels less than or equal to that of patients 49
  • 58. undergoing deep brain stimulation, which carries a lower morbidity risk (Lane et al., 2008). Cell transplantation: Transplantation of primary ventral mesencephalic tissue into the striatum aims to restore brain circuitry and function lost as a result of PD. The main objective of primary tissue transplantation has been to provide proof of principle that grafted dopaminergic neurons can i) survive and restore regulated dopamine release, ii) integrate with the host brain to reinstate frontal cortical connections and activation, and iii) lead to measurable clinical benefits together with improved quality of life. Preclinical work in animal models of PD has shown that grafted dopaminergic neurons, extracted from the developing ventral mesencephalon (VM) can survive, reinnervate the lesioned striatum, and improve motor function (Winkler et al 2005). Over the past two decades, a series of open label clinical trials have provided convincing evidence to show that human embryonic nigral neurons taken at a stage of development when they are committed to a dopaminergic phenotype can survive, integrate and function over a long time in the human brain. There is good evidence of graft survival, with grafted neurons developing afferent and efferent projections with the host neurons. Long term survival of dopaminergic grafts is possible up to 10 years after transplantation, and there have been no 50
  • 59. reported cases of overt immunorejection even after several years of withdrawal from immuno-suppression (Olanow et al 2003). Evidence from Positron Emission Tomography (PET) scanning has revealed significant increases in activation in the areas reinnervated by the grafted cells, and longitudinal clinical assessments indicate significant functional recovery for motor control, in some cases for more than 10 years, in the most successful cases, patients have either reduced dependency for or completely withdrawn from L-dopa treatment. Post mortem studies similarly show good survival of transplanted neurons and well integrated grafts (figure 16) (Winkler et al., 2005). Figure (16): Dopamine Neuron Transplantation: PET images from a Parkinson’s patient before and after fetal tissue transplantation, the image taken before surgery (left) shows uptake of a radioactive form of dopamine (red) only in the caudate nucleus, indicating that dopamine neurons have degenerated. Twelve months after surgery, an image from the same patient (right) reveals increased dopamine function, especially in the putamen (Winkler et al., 2005). 51
  • 60. The precise mechanism responsible for these dyskinesia remains unknown but it does not appear to be related to graft overgrowth resulting in excessive dopamine release. One possibility surrounds the quality of dissected tissue. Successful trials have used either freshly dissected tissue or tissue that has been stored in culture for only a few days. One of the trials reporting cases of severe dyskinesias used tissue stored in culture for up to four weeks and it may be that holding tissue in this way reduces its dopaminergic composition (Olanow et al., 2003). A further issue concerns the identification of dense hyperdopaminergic areas within the graft of some patients with graft induced dyskinesias. This may have caused uneven striatal innervation and excessive dopamine release into non reinnervated areas. It is also possible that variable side effects of graft induced dyskinesias are related to patient selection. Greater functional improvement is associated with younger patients, and in patients with less advanced disease. This is most likely because the neuropathology is relatively confined to the nigrostriatal pathway and may have better trophic support compared to patients with more advanced disease (Winkler et al., 2005). Stem cells: Stem cells could provide one such source and would overcome the issue of limited availability of fresh primary fetal cells. A wide range of stem cells are being investigated as 52
  • 61. potential sources of dopaminergic neurons for transplantation, stem cells can be obtained from various sources (Morizane et al., 2009). The majority of research thus far with respect to the formation of dopaminergic neurons for the treatment of PD is in embryonic stem cells and neural stem cells. Dopaminergic neurons are more easily obtained from neural stem cells in the developing ventral mesencephalon (VM) than other parts of the developing central nervous system but the number of dopaminergic cells produced is still very low. Despite genetic manipulation and the addition of various growth and differentiation factors, generating large numbers of dopaminergic cells from this cell type has had mixed results (Panchision, 2006). However, greater success has been achieved with the more complex ES cells. Derived from blastocysts donated following in vitro fertilization these cells are truly pluripotent. Promising data have been obtained with dopaminergic neurons derived from mouse ES cells, significantly improving motor function in a rat model of PD. However, directing the differentiation of human ES cells has proved complex and while 50% of cells spontaneously differentiate into neurons upon leukemia inhibitory factor (LIF) withdrawal, few are dopaminergic. Thus, there is the need to develop protocols to ‘direct’ differentiation. The most successful published protocols describe multiple culture stages in which different transcription and growth 53
  • 62. factors are added at controlled time points (Goldman and Windrem, 2006). However, despite good yield of dopaminergic neurons in vitro, clinically relevant long term survival and behavioral recovery in animal models rivaling that of primary tissue has yet to be convincingly demonstrated. Neuronal stem cells unlike embryonic stem cells, which are only derived from the embryonic blastocyst, neural stem cells can be found both in embryonic neural tissue and also in specific neurogenic regions of the adult brain. If the in vivo survival of neural stem cells can be improved they hold the potential to provide autologous transplantation as patients provide the cells for their own recovery (Morizane et al., 2009). Interestingly, stem cells may not just be useful as dopamine factories in the striatum. Some studies in both rodent and primate models have shown significant behavioral recovery following transplantation with neural stem cells. In addition to the generation of a small population of dopaminergic neurons other cells within the graft were found to be releasing growth factors which are purportedto exert neuro-protective or neuroregenerative influences. While more evidence needs to be accumulated on the longevity of this effect, it broadens the potential of neural stem cells from simple dopamine replacement to preserving and enhancing remaining dopaminergic neurons (Svendsen and Langston, 2004). 54
  • 63. Stem cell based approaches could be used to provide therapeutic benefits in two ways: first, by implanting stem cells modified to release growth factors, which would protect existing neurons and/or neurons derived from other stem cell treatments; and second, by transplanting stem cell derived DA neuron precursors/neuroblasts into the putamen, where they would generate new neurons to ameliorate disease-induced motor impairments (figure 17) (Lindvall and Kokaia, 2010). Figure (17): Stem cell based therapies for PD. PD leads to the progressive death of DA neurons in the substantia nigra and decreased DA innervation of the striatum, primarily the putamen (Lindvall and Kokaia, 2010). Neurogenesis: As mentioned above endogenous stem cells are present in specific regions of the brain. While the occurrence of neurogenesis in the striatum and substantia nigra is debated, one 55
  • 64. indisputable neurogenic region is the subventricular zone (SVZ) lying adjacent to the striatum. The cells in the region are an assortment of stem and progenitor cells that have the potential to be mobilized and induced to differentiate by the presence of growth factors or other small molecules. In the normal condition 75%–99% or the cells differentiate into granular GABAergic neurons, with the rest forming periglomular neurons expressing either tyrosine hydroxylase or GABA. The control of proliferation and mobilisation of these cells may be dopaminergic as both MPTP and 6-OHDA mediated dopamine depletion reportedly decreases proliferation in this zone (Zhao et al., 2008). An additional source of endogenous source of new dopaminergic neurons may be described presence of tyrosine hydroxylase positive cell bodies in the striatum, which increase in quantity with dopaminergic denervation. As yet there are no imminent therapeutic strategies heading towards the clinic that manipulate these endogenous systems but their potential is waiting to be harnessed. Therapeutic strategies to increase striatal dopamine could involve recruiting newly produced neurons in the SVZ and encouraging them to migrate into the striatum and differentiate into dopaminergic neurones or to stimulate cells resident in the striatum. In order for this to be achieved understanding more about these two processes of neurogenesis and phenotypic switching in the striatum is necessary, determining the intrinsic or extrinsic factors 56
  • 65. responsible may provide an alternative set of mechanisms that could be utilized to treat PD (Lane et al., 2008). Graft standardization: Grafting methods for the past 20 years have differed in everything from procurement process to tissue composition to implantation technique. To add even more variability, multiple donors are needed to create a graft large enough to carry some promise of efficacy. This, undoubtedly, plays an important role in determining survival, growth, and integration of the transplant (Morizane et al., 2009). As stem cells can theoretically provide an endless source of quality consistent neurons, standardization of the transplant tissue will enhance the reliability of the procedure and its results. One promising study has shown that implantation of undifferentiated human neural stem cells (hNSCs) in Parkinsonian primate brains can restore functionality. Furthermore, the repair process not only reestablishes the gross anatomical structure of the organ but does so with appropriate proportions of neuron types. Guided by signals of the microenvironment of the damaged brain, uncommitted hNSCs are induced to differentiate into dopaminergic neurons, as well as other cells that mediate neuroprotection (Lane et al., 2008). Patient standardization: With standardization of transplant material, patients must likewise be evaluated for variables in their presentation of the disease. Specifically, the distribution of the damaged neurons 57
  • 66. should be taken into account before and after graft implantation. In earlier studies, patient selections overlooked the preoperative magnitude of the lesions, making it difficult to evaluate the extent of the graft incorporation. Similarly, it was also unknown whether continued postoperative degeneration of non grafted regions would affect clinical response. Conversely, patients with little or no postoperative damage showed the best functional outcome. Because the decline of dopaminergic cells in areas outside the nigrostriatal region seems to arise as PD progresses to a more severe state, it may be that implantation during earlier stages will exhibit a higher rate of success. This is mirrored in the survival of transplanted tissue, which survives and integrates better in younger patients. The reasons why this occurs have yet to be fully determined, but it is known that neural growth factors are expressed more in younger brains (Morizane et al., 2009). 58
  • 67. Stem cell therapy in stroke Stroke is defined as an abrupt focal loss of brain function resulting from interference with the blood supply to part of the central nervous system. It is one of the major causes of death and disability among the adult population in the world. In spite of the extensive research in the field of stroke biology, there is little effective treatment for a completed stroke. Most strokes fall into two main categories: ischemic (80%) or hemorrhagic (20%) (Caplan, 2011). The biology of cellular transplantation: The transplantation of human neuronal cells is an approach to reducing the functional deficits caused by CNS disease or injury. Several investigators have evaluated the effects of transplanted fetal tissue, rat striatum, or cellular implants into small animal stroke models for the most part, clinical trial designs using primary human fetal tissue into patients with neurodegenerative diseases have lessened. The widespread clinical use of primary human tissue is likely to be limited due to the ethical and technical difficulties in obtaining large quantities of fetal neurons at the same time; much effort has been devoted to developing alternate sources of human neurons for use in transplantation (Kondziolka and Wechsler, 2008). When transplanted, these neuronal cells survived, extended processes, expressed neurotransmitters, formed functional synapses, and integrated with the host. During the retinoic acid 59
  • 68. induction process, significant changes were seen in the neuron precursor cells that resulted in the loss of neuroepithelial markers and the appearance of neuronal markers. The final cell product was a ≥ 95% pure population of human neuronal cells that appeared virtually indistin-guishable from terminally differentiated, post mitotic neurons. The cells were capable of differentiation to express a variety of neuronal markers characteristic of mature neurons, including all 3 neurofilament proteins (L, M, and H); microtubule associated protein 2, the somatic/dendritic protein, the axonal protein. Thus, the neuronal phenotype made these cells a promising candidate for replacement in patients with CNS disorders, as a virtually unlimited supply of pure, post mitotic, and differentiated human neuronal cells (Lindvall and Kokaia, 2010). The concept of restoring function after a stroke by transplanting human neuronal cells into the brain was conceived in the mid 1990s. Research conducted in a rat model of transient focal cerebral ischemia demonstrated that transplantation of fetal tissue restored both cognitive and motor functions. Ischemic stroke leads to the death of multiple neuronal types and astrocytes, oligodendrocytes, and endothelial cells in the cortex and subcortical regions. Stem cell based therapy could be used to restore damaged neural circuitry by transplanting stem cell derived neuron precursors/neuroblasts. Also, compounds could be infused that would promote neurogenesis from endogenous SVZ stem/progenitor cells, or stem cells could be 60
  • 69. injected systemically for neuroprotection and modulation of inflammation (Kondziolka and Wechsler, 2008). Behavioral testing was conducted using a passive avoidance learning and retention task and a motor asymmetry measure. Animals that received transplants of neurons and treatment with cyclosporine showed amelioration of ischemia induced behavioral deficits throughout the 6 month observation period. They demonstrated complete recovery in the passive avoidance test, as well as normalization of motor function in the elevated body swing test. In comparison, control groups receiving transplants of rat fetal cerebellar cells, medium alone, or cyclosporine failed to show significant behavioral improvement. Subsequent studies have shown that these cells released glial derived neurotrophic factor after transplantation into ischemic rats (figure 18) (Lindvall and Kokaia, 2010). Figure (18): Stem cell based therapies for stroke (Lindvall and Kokaia, 2010). 61
  • 70. Embryonic stem cells: Animal models have demonstrated that ESCs, when transplanted into adult hosts, differentiate and develop into cells and tissues and thus may be applicable for treating a variety of conditions, including Parkinson’s disease, multiple sclerosis, spinal injuries, stroke, and cancer. Transplanted ESCs are exposed to immune reactions similar to those acting on organ transplants; hence, immunosuppression of the recipient is generally required. It is possible, however, to obtain ESCs that are genetically identical to the patient’s own cells using therapeutic cloning techniques (Kalluri and Dempsey, 2008). Several studies showed that these cells are able to migrate in response to damage, using MRI, that ESCs that were implanted into the healthy hemisphere of rat brains 2 weeks after focal cerebral ischemia (FCI), migrated along the corpus callosum to the ventricular walls, and populated en masse at the border zone of the damaged brain tissue (i.e., the hemisphere opposite to the implantation sites). Another study showed that undifferentiated ESCs xenotransplanted into the rat brain at the hemisphere opposite to the ischemic injury migrated along the corpus callosum toward the damaged tissue and differentiated into neurons at the border zone of the lesion. In the homologous mouse brain, the same murine ESCs did not migrate, but produced highly malignant teratocarcinomas at the site of implantation, independent of whether they were predifferentiated in vitro to NPCs. These results imply that 62
  • 71. ESCs might migrate to the damaged site. However, the production of teratocarcinoma raises concerns about the safety of ESC transplantation in patients with stroke (Battler and Leor, 2006). Adult neural stem cells: During the last century, the dogma existed that the adult CNS was incapable of generating new neurons (neurogenesis). Over the past decades, convincing evidence emerged that neurogenesis in the adult CNS is a continuous physiological process. Neurogenesis is present in two regions: the subventricular zone (SVZ) and the subgranular zone of the dentate gyrus (figure 19) (Kalluri and Dempsey, 2008). Figure (19): Schematic drawings showing ischemia induced damage in the cortex and neural stem cell proliferation in the subventricular zone and dentate gyrus; these hypothetical sections of brain illustrate the ischemic tissue and the neurogenic regions. A: Drawing showing the subventricular zone of the lateral ventricles. B: Drawing showing the dentate gyrus in the hippocampus. Note the migration of cells from the subventricular zone toward the infarcted area. Red dots represent proliferating and/or migrating neural stem and/or progenitor cells (Kalluri and Dempsey, 2008). 63
  • 72. Additionally, other studies also indicated the existence of NSCs in other regions of the CNS, namely the striatum, spinal cord and neocortex. SVZ and dentate gyrus derived NSCs are characterized by long term, self renewal capacity and multipotency. Adult SVZ and dentate gyrus derived NSCs persist throughout the life span of mammals including humans. It is important to note that neurogenesis occurs in a physiological mode or is exogenously modulated by external signals or pathophysiological processes. External global stimulants such as enriched environment, physical activity and stress or application of defined molecules such as fibroblast growth factor-2, vascular endothelial growth factor (VEGF), brain derived neurotrophic factor (BDNF) and erythropoietin differentially modulate adult neurogenesis. Finally, CNS disease conditions such as seizures and traumatic brain injury represented by respective animal models induce neurogenesis (Panchision, 2006). Role of neural stem cells: Proliferation: Neural stem cell proliferation involves the sequential activation of several cell cycle dependent enzymes and proteins to initiate either symmetrical (2 stem cells) or asymmetrical cell division (1 stem and 1 progenitor cell). Erroneous activation of cell cycle enzymes in differentiated cells like neurons, however, can lead to either apoptotic death of the neurons or to the formation of cancer cells. A plethora of growth factors are 64
  • 73. expressed by the ischemic tissue, which may be responsible for the ischemia induced neurogenesis. The increase in ischemia induced neurogenesis could therefore be due to the upregulation of growth factor content or their receptor expression (Hass et al., 2005). In addition, not all growth factors are stimulatory in function. Although a variety of growth and trophic factors are upregulated following ischemic injury, some are stimulators of neural progenitor proliferation, whereas others block the self renewal of cells. It is important to note that ischemia induced migration of neuroblasts has been shown to be due to the expression of monocyte chemoattractant protein (MCP-1), which can attract the progenitor cells away from the neurogenic niche, blocking their proliferation. Likewise, inhibition of growth factor activity by its binding proteins may also interfere with proliferation of cells. Interestingly, insulin like growth factor–I (IGF-I) can stimulate the proliferation of progenitor cells only in the presence of mitogens like fibroblast growth factor–2 (FGF-2) and promotes differentiation following mitogen (FGF-2) withdrawal (figure 20) (Kalluri and Dempsey, 2008). 65
  • 74. Figure (20): Schematic drawing showing proliferation and differentiation of neural stem cells. These cells proliferate in response to mitogens and differentiate into neurons, astrocytes, and oligodendrocytes on exposure to various growth factors (Kalluri and Dempsey, 2008). Migration: Several studies have shown that ischemia induces the migration of neuroblasts into the striatum and cerebral cortex. An enriched environment, however, increased the stroke induced neurogenesis in the hippocampus, but decreased the cell genesis and migration of neuroblasts into striatum. In addition, erythropoietin has also been shown to be involved in the proliferation of progenitor cells in the subventricular zone. Hence, it appears that proliferation and migration of cells may be interrelated, in the sense that they are elements of the same process (Son et al., 2006). 66
  • 75. It is thought that chemokines proteins released at the site of injury by the inflammatory cells induce migration of the cells. The expression of chemokine receptors on neural stem cells is important for an efficient response to chemokines. The expression of chemokine receptors is regulated by retinoic acid, and that animals fed on a diet lacking retinoic acid have decreased numbers of double cortin positive cells. Retinoic acid has been shown to induce the differentiation of neurons in vitro. Hence, it appears that lack of retinoic acid may have decreased the neuronal differentiation and/ or the expression of chemokine receptors, which results in decreased numbers of neuroblasts (immature neuronal cells expressing double cortin) migrating out of the neurogenic niche (figure 21) (Kondziolka and Wechsler, 2008). 67
  • 76. Figure (21): Schematic drawing showing ischemia induced neurogenesis. Ischemia increases the expression of IGF-I, FGF- 2, TGFb1, and MCP-1. The IGF-I and FGF-2 enhance the proliferation of cells, whereas MCP-1 increases the migration of cells away from neurogenic regions toward the ischemic tissue. The stem cells leaving the neurogenic regions exit the cell cycle, during which time TGFb1 promotes the differentiation of stem cells into neuroblasts (migrating immature neurons). Alternately, if proliferating cells in the neurogenic region are exposed to TGFb1, it will inhibit the process, thus slowing down the proliferation of cells (Kondziolka and Wechsler, 2008). Differentiation: The process of generating specialized cells from neural stem cells is called differentiation. Neural stem cells can be differentiated into neurons, astrocytes, and oligodendrocytes; this involves interplay between intrinsic cellular programs and extrinsic cues like growth factors provided by the surrounding environment. Although neural progenitor cells proliferate in response to ischemia, several immature neurons (double cortin positive cells) were shown to be migrating toward striatum and cortex (Battler and Leor, 2006). 68
  • 77. Although IGF-I induces the proliferation and differentiation of progenitor cells after ischemia, most of the cells that are migrating toward the injury site are neuroblasts (immature neurons) but not oligodendrocytes. Although speculative, a neuronal differentiation factor like transforming growth factor b1 (TGFb1), which is upregulated after ischemia, may be responsible for the formation of neuroblasts during the post ischemic neurogenesis. The differential response of IGF-I toward proliferation and differentiation was shown to be regulated by a mitogen activated protein kinase pathway. Because matrix metalloproteinase (MMPs) play an important role in both extracellular matrix (ECM) digestion and IGF regulation, it is crucial to understand their direct (ECM digestion) and indirect (IGF metabolism) effects on progenitor cell differentiation. Hence it is possible that after stroke, cells proliferate in response to mitogens and growth factors, some of which exit the cell cycle due to the chemokine mediated migration out of the neurogenic area, and differentiate into neuroblasts in response to differentiation promoting factors such as TGFb1. Thus, the final outcome depends on the spatial and temporal expression of these factors following ischemia (Son et al., 2006). Potential mechanism involved in stem cell mediated recovery after stroke: Initial transplantation studies were focused on the potential of NSCs to replace lost circuitry. Transplanted neural progenitor 69
  • 78. cell (NPCs) in a rat model of global ischemia have been reported to express synaptic proteins post transplantation. However, only limited evidence demonstrates that transplanted cells are able to sustain CNS repair through massive cell replacement, especially to the extent that might be required after stroke. Regardless of the characteristics of the experimental disease, functional recovery achieved through NPC transplantation does not correlate with absolute numbers of transplant derived, newly generated, and terminally differentiated neural cells (Emerson et al., 2008). Stem cell induced neuroprotection: Transplanted stem cells can provide neuroprotection and reduce host cell death in the post stroke brain. Most authors have reported functional recovery and a reduction in lesion size when cells are transplanted within the first 24-48 hours after stroke. The short timeframe in which NPCs affect recovery cannot be explained by the regeneration of new neurons and synapses, suggesting an important role for neuroprotection in enhancing recovery. In fact, NPCs are known to exert direct neuroprotection through the neutralization of free radicals, inflammatory cytokines, excitotoxins, lipases, peroxidases, and other toxic metabolites that are released following an ischemic event (Ourednik et al., 2005). The neuroprotective effects of transplanted NPCs are usually accompanied by increased in vivo bioavailability of main neurotrophins such as nerve growth factor, brain derived 70
  • 79. neurotrophic factor (BDNF), ciliary neurotrophic factor, vascular endothelial growth factor (VEGF), fibroblast growth factor, and glial derived neurotrophic factor (GDNF). Alternatively, one could argue that the cells did adhere to the endothelium in the affected brain area and exerted their effect through the secretion of GDNF but did not eventually engraft. A different aspect of neuroprotection would be an effect on endogenous neurogenesis after stroke. Endogenous neurogenesis is increased after a stroke. Its exact function has yet to be determined, but it may signify a natural repair mechanism of the brain, which could be enhanced by transplanted cells (Guzman et al., 2008). Stem cell induced angiogenesis: Transplanted NPCs can also enhance endogenous angiogenesis. Increased vascularization in the penumbra within a few days after stroke is associated with spontaneous functional recovery. As early as 3 days after ischemic injury, new blood vessels are observed in the stroke affected penumbra, and growth continues to increase for at least 21 days. Transplanted cell induced blood vessel formation has been reported with BMSCs, NSCs, and cells from human cord blood or peripheral blood. The ability of transplanted cells to increase endogenous levels of angiogenic factors and chemoattractant factors (for example, SDF-1) induces the proliferation of existing vascular endothelial cells (angiogenesis) and mobilization and homing of 71
  • 80. endogenous endothelial progenitors (vasculogenesis) (Shyu et al., 2006). The direct injection of recombinant stromal derived factor–1 (SDF-1) into the stroke affected rat brain resulted in the increased recruitment of BMSCs and increased vascular density in the ischemic brain. A similar effect was achieved by treatment of stroke affected rats with granulocyte colony stimulating factor (Ourednik et al., 2005). Stem cell induced immunomodulation: In addition to neuroprotection and enhancement of angiogenesis, transplanted NPCscan also decrease post ischemic inflammatory damage. A landmark study highlights the remarkable ability of transplanted NPCs to promote neuroprotection through an immunomodulatory strategy, once within inflamed CNS areas, systemically injected NPCs persist around the perivascular space where reactive astrocytes, inflamed endothelial cells, and bloodborne infiltrating T cells coreside. Adult NPCs can promote neuroprotection by releasing antiinflammatory chemokines and by expressing immunomodulatory molecules (Pluchino et al., 2005). In animal models of stroke, decreased infiltration of mononuclear cells has been demonstrated at the lesion borders of ischemic areas in the CNS where NPCs accumulate. Working under the paradigm of bidirectional communication between transplanted cells and the inflammatory ischemic microenvironment, one group has attempted to combine NPC 72