ArticlePDF AvailableLiterature Review

Neuroprotective Effect of Granulocyte-Colony Stimulating Factor

Authors:

Abstract and Figures

Granulocyte-colony stimulating factor (G-CSF) is a growth factor which stimulates proliferation, differentiation, and survival of hematopoietic progenitor cells. G-CSF is being used extensively in clinical practice to accelerate recovery of patients from neutropenia after cytotoxic therapy. However, growing evidences have suggested that G-CSF has important non-hematopoietic functions in central nervous system. Recent studies have shown the presence of G-CSF/G-CSF-receptor (G-CSFR) system in the brain, and their roles in neuroprotection and neural tissue repair as well as improvement in functional recovery. The increased expression of G-CSF/G-CSFR on neurons subjected to hypoxia provides evidence that G-CSF may have an autocrine protective signaling mechanism in response to neural injury. G-CSF exerts neuroprotective actions through the inhibition of apoptosis and inflammation and the stimulation of neurogenesis. Moreover, G-CSF has been shown to mobilize bone marrow stem cells into the injured brain improving neural plasticity. In this review, we summarize some of the recent studies on G-CSF and the corresponding signal transduction pathways regulated by G-CSF in neuroprotection.
Content may be subject to copyright.
[Frontiers in Bioscience 12, 712-724, January 1, 2007]
712
Neuroprotective Effect of Granulocyte-Colony Stimulating Factor
Ihsan Solaroglu
1
, Vikram Jadhav
1
, and John H. Zhang
1,2,3
1
Department of Physiology and Pharmacology,
2
Division of Neurosurgery,
3
Department of Anesthesiology, Loma Linda
University School of Medicine, Loma Linda, CA
TABLE OF CONTENTS
1. Abstract
2. Introduction
3. G-CSF and its receptor
3.1. G-CSF and G-CSFR in the brain
4. Clinical applications of G-CSF
4.1. G-CSF in general clinical use
4.2. G-CSF treatment in patients with cerebral injuries
5. Neuroprotective properties of G-CSF
5.1. G-CSF, inflammation, and apoptosis
5.2. G-CSF, hematopoietic stem cell mobilization, and brain repair
5.3. G-CSF and neurogenesis
6. Summary and Perspective
7. Acknowledgement
8. References
1. ABSTRACT
Granulocyte-colony stimulating factor (G-CSF) is
a growth factor which stimulates proliferation,
differentiation, and survival of hematopoietic progenitor
cells. G-CSF is being used extensively in clinical practice
to accelerate recovery of patients from neutropenia after
cytotoxic therapy. However, growing evidences have
suggested that G-CSF has important non-hematopoietic
functions in central nervous system. Recent studies have
shown the presence of G-CSF/G-CSF-receptor (G-CSFR)
system in the brain, and their roles in neuroprotection and
neural tissue repair as well as improvement in functional
recovery. The increased expression of G-CSF/G-CSFR on
neurons subjected to hypoxia provides evidence that G-
CSF may have an autrocrine protective signaling
mechanism in response to neural injury. G-CSF exerts
neuroprotective actions through the inhibition of apoptosis
and inflammation and the stimulation of neurogenesis.
Moreover, G-CSF has been shown to mobilize bone
marrow stem cells into the injured brain improving neural
plasticity. In this review, we summarize some of the recent
studies on G-CSF and the corresponding signal
transduction pathways regulated by G-CSF in
neuroprotection.
2. INTRODUCTION
Each year about 700,000 people experience a
new or recurrent stroke which is the leading cause of adult
disability and remains the third most common cause of
death in the United States (1). For the last two decades, a
great deal of attention has been paid to identify the
pathophysiological pathways leading to neural death in
stroke and to design neuroprotectant agents to modulate
these pathways. Although numerous agents have been
found to reduce infarct size and improve clinical outcome
in experimental models, the clinical use of these
neuroprotective agents has been hampered by toxicity of
the compounds. Recombinant tissue plasminogen activator
(rt-PA) remains the only approved agent for acute stroke
management in humans. However, its narrow therapeutic
window and potential side effects including increased risk
of intracerebral
hemorrhage and neurotoxicity limit the
efficacy of rt-PA (2-4).
In recent years, several independent research
groups have reported neuroprotective properties of growth
factors such as erythropoietin (EPO), insulin-like growth
factor, basic fibroblast growth factor, brain-derived
neurotrophic factor (BDNF), and granulocyte-colony
Neuroprotection by G-CSF
713
stimulating factor (G-CSF) in central nervous system
diseases such as stroke, neurotrauma, neuroinflammatory,
and neurodegenerative diseases using experimental animal
models (5-12). G-CSF, a member of growth factor family, not
only stimulates the development of committed progenitor cells
to neutrophils but also modulates the neutrophil functions and
their distribution in the body (13). Furthermore, like other
cytokines, it also has trophic effects on the different cell types
including neuronal cells (14). This could be of interest, because
neurotrophic factors are not only essential for the survival and
differentiation of normally developing neurons, but they also
play important roles in the protection and recovery of mature
neurons under pathologic conditions (15). Although there is
evidence from recent experimental studies that administration
of G-CSF is neuroprotective (16-20), the precise mechanisms
of the neuroprotective effect of G-CSF are not entirely
explored.
In this article, we review the biological properties
and clinical applications of G-CSF, and the possible role
and novel mechanisms of G-CSF as a potential therapeutic
agent for neuroprotection.
3. G-CSF AND ITS RECEPTOR
G-CSF is a glycoprotein with a molecular mass
of 19 kDa and is structurally characterized by four anti-
parallel alpha-helices (21). G-CSF is produced by a variety
of cells including bone marrow stromal cells, endothelial
cells, macrophages, fibroblasts, and astrocytes in response
to specific stimulation (13, 22, 23). Human G-CSF is
encoded by a single gene that is located on chromosome 17
q11-12 (24, 25).
G-CSF binds to its specific receptor and
stimulates the proliferation and differentiation of
neutrophilic progenitor cells. The G-CSF receptor (G-
CSFR) is a type I membrane protein and has a composite
structure consisting of an immunoglobulin-like domain, a
cytokine receptor-homologous domain and three
fibronectin type III domains in the extracellular region (26).
G-CSFR is expressed not only on a variety of
hematopoietic cells including neutrophils, and their
precursors, monocytes, platelets, lymphocytes, and
leukemia cells, but also on non-hematopoietic cells such as
endothelial cells, neurons and glial cells (20, 27-32).
G-CSFR activates a variety of intracellular
cascades, including the Janus
kinase (JAK)/signal
transducer and activator of transcription (STAT), the
Ras/mitogen-activated
protein kinase (MAPK), and
phosphatidylinositol 3-kinase (PI3-K)/protein kinase B
(PKB) (also known as Akt) (33-36). Activation of these
cascades subsequently activates their downstream
substrates and affects target genes which mediate
proliferation, differentiation, and survival of hematopoietic
cells (37). These signaling cascades are summarized in
Figure 1.
3.1. G-CSF and G-CSFR in the brain
G-CSF receptor and its ligand have been shown
to be expressed by neurons in a variety of brain regions
including pyramidal cells in cortical layers (particularly in
layers II and V), Purkinje cells in the cerebellum,
subventricular zone (SVZ) and cerebellar nuclei in rats. G-
CSF positive cells in CA3 region of the hippocampus,
subgranular zone and hilus of the dentate gyrus, entorhinal
cortex, and olfactory bulb have been identified (12).
Moreover, G-CSF receptor expression has shown in the
frontal cortex of human brain by postmortem studies (12).
G-CSF and its receptor are co-expressed in
neurons in the rodent central nervous system and are
upregulated in the ipsilateral forebrain hemisphere after 2
hours occlusion of the middle cerebral artery (MCAO) and
reperfusion (12). Similarly, Kleinschnitz et al. showed that
4 hours after permanent MCAO, G-CSF mRNA levels
massively increases compared to normal cortex and
decreases after 2 days to its control levels (38). The authors
pointed out that the increase in G-CSF mRNA expression is
not only within the ischemic lesions but also in the
nonischemic frontal cortex after photothrombosis model of
focal cerebral ischemia (38). Taken together, current
evidences suggest that G-CSF may have an autrocrine
protective signaling mechanism in response to neural
injury. Similar mechanisms have been suggested for other
growth factors, especially EPO which have been
extensively reviewed by others (39, 40).
4. CLINICAL APPLICATIONS OF G-CSF
4.1. G-CSF in general clinical use
G-CSF, specifically Filgrastim (r-metHuG-CSF),
a genetically engineered drug was approved by FDA on
February 21, 1991 to decrease the incidence of infection, as
manifested by febrile neutropenia, in patients with
nonmyeloid malignancies receiving myelosuppressive
anticancer drugs associated with a significant incidence of
severe neutropenia with fever. During the last decade, G-
CSF has found wide spread use in reducing the duration of
febrile neutropenia in cancer patients treated with
chemotherapy. G-CSF is also being used clinically to
facilitate hematopoietic recovery after bone marrow
transplantation, the mobilization of peripheral blood
progenitor cells in healthy donors and in the treatment of
severe congenital neutropenia (41-44).
The role of G-CSF treatment in variety of other
conditions is still matter of discussion. There is evidence
from clinical studies that administration of G-CSF is safe,
beneficial, and well
tolerated for the prevention or
treatment of infections in patients with
non-neutropenic
infections, patients undergoing surgery, patients with
human immunodeficiency virus, and patients with diabetic
foot infections (45-50). On the other hand, controlled trials
and systematic reviews have concluded that there is no
current evidence supporting the routine use of G-CSF in the
treatment of pneumonia, in patients undergoing surgery, for
treating or preventing neonatal infections, and in the
treatment of diabetic foot infections (51-54).
4.2. G-CSF treatment in patients with cerebral injuries
The prophylactic administration of G-CSF
against severe infections in critically ill patients with
Neuroprotection by G-CSF
714
Figure 1. Figure shows the hematopoetic actions of Granulocyte-Colony Stimulating Factor (G-CSF). G-CSF plays an important
role in the survival, proliferation and differentiation of the hematopoetic cells. The figure also depicts the signaling pathways
suggested in the existing literature for the G-CSF mediated actions. G-CSFR activates Janus
kinase (JAK)/signal transducer and
activator of transcription (STAT), the Ras/mitogen-activated
protein kinase (MAPK), and phosphatidylinositol 3-kinase (PI3-
K)/Akt pathways. Activation of JAK2/STAT3 leads
to increased expression of cellular inhibitor of apoptosis protein2 (cIAP2) in
human neutrophils resulting in their survival. Activation of STAT3 by G-CSF has also been linked to myeloid cell differentiation.
STAT5, however, seems to be involved in G-CSF-dependent cell
proliferation.
Neuroprotection by G-CSF
715
cerebral injuries is controversial. The primary outcome of
studies is a reduction in frequency of bacteremia and in
life-threatening infections in patients with cerebral injuries.
For the first time, Heard et al. conducted a randomized,
placebo-controlled, double-blind, multi-center, phase II
study to determine whether the use of prophylactic
recombinant human G-CSF reduces the frequency of
nosocomial infections in patients with either acute
traumatic brain injury or cerebral hemorrhage (55). The
primary efficacy end points of this study were the
frequency of nosocomial infections, increase in absolute
neutrophil count, and the safety of G-CSF. Secondary
efficacy end points included serum G-CSF concentrations,
28 day survival, and duration of hospitalization, antibiotic
use, intensive care unit stay, and mechanical ventilation.
Authors reported that G-CSF increases absolute neutrophil
counts and markedly reduces the frequency of bacteremia
in a dose-dependent fashion in critically ill and intubated
patients (55). They concluded that phase III trial is
warranted to confirm these results. However, the authors
did not report neurological outcomes and cerebral
physiological variables of the patients. Also, the study was
criticized by Whalen et al. because the systemic
administration of G-CSF to patients with traumatic brain
injury may carry potential risk of increased acute
inflammatory reactions in the injured brain due to excessive
accumulation of neutrophils (56). Whalen et al. have
suggested that it was premature to use G-CSF in humans
with TBI until animal studies have demonstrated G-CSF
effects on brain injury after trauma or stroke (56).
Combined therapy with high-dose barbiturates
and mild hypothermia has been used widely for patients
with severe head injuries to decrease their intracranial
pressure (ICP). However, both therapies alone may result in
a decrease in leukocytes and suppression of neutrophil
functions which increase susceptibility to infectious
complications (57, 58). Ishikawa et al. have used G-CSF to
ameliorate life-threatening infections in patients with
severe head injury who were treated with combined therapy
involving high-dose barbiturates and mild hypothermia
(59). Authors studied the daily changes in total leukocyte
count, leukocyte differentiation, C-reactive protein,
respiratory index (RI), and ICP. Patients treated with G-
CSF showed significant improvement in the RI which
reflects the recovery from pneumonia. Authors reported
that ICP was not elevated by G-CSF administration, and a
significant improvement was achieved in survival rate.
Moreover, G-CSF treatment resulted in decrease of blood
IL-6 concentrations of patients. Although previous studies
that have attempting to correlate IL-6 production
with
patient outcome following TBI have produced inconsistent
results, Arand et al. suggested that elevated IL-6 in the
serum
or CSF is associated with poor outcome (60).
Ishikawa et al. concluded that G-CSF treatment ameliorates
life-threatening infections in patients with severe head
injury who were treated with combined therapy involving
high-dose barbiturates and mild hypothermia without
causing lung injury or increasing brain swelling (59).
However, this was not a prospective study and the number
of patients was small to conclude whether the use of G-CSF
in brain injured patients is a safe prophylactic agent against
severe infections.
In summary, the therapeutic potential of
prophylactic G-CSF therapy in critical ill patients, and G-
CSF administration for the prevention or treatment of
infections in non-neutropenic patients are currently
unproven and further controlled trials will be necessary to
clarify these issues.
5. POTENTIAL NEUROPROTECTIVE
PROPERTIES OF G-CSF
There is growing number of studies about the
neuroprotective properties of growth factors in the last
decade. Although G-CSF has been used for many years in
clinical practice and is a well known growth factor, there
are only a few experimental studies exploring the
neuroprotective effects of G-CSF. These studies are
summarized in Table 1.
5.1. G-CSF, inflammation, and apoptosis
It is well established that the inflammation, in
response to brain injury, involves infiltration of neutrophils
and monocytes/macrophages into the injured brain
parenchyma and activation of resident brain cells which are
capable of generating inflammatory mediators (61, 62).
Although there is not a clear cause-effect relationship
between neutrophil recruitment and CNS pathogenesis
(63), it is well known that neutrophil activation results in
release of proteolytic enzymes and generation of free
oxygen radicals. Excessive generation of free oxygen
radicals causes DNA damage, lipid peroxidation and
inactivation of proteins and finally leads to severe tissue
injury (64-66). Free oxygen radicals also contribute to the
breakdown of the blood-brain barrier (BBB) and brain
edema (62, 67). It could be of interest because G-CSF
administration increases absolute neutrophil count in
peripheral blood which may result in acute inflammatory
reactions in the injured brain. In support of this, it has been
reported that in vivo chloroquine or colchicine treatment
that reduce the number of mononuclear phagocytes in
damaged brain, help to block reactive astrogliosis and
neovascularization, and slow the rate of debris clearance
from sites of traumatic injury (68). For the first time,
Whalen et al. evaluated the effect of G-CSF-induced
neutrophilia on BBB permeability and brain edema after
traumatic brain injury in a rat model (69). They have
demonstrated that subcutaneous administration of G-CSF
(25 µg/kg every 12 hrs for five doses, with the last dose
administrated immediately before cortical impact injury
(CCI)) resulted in increased systemic absolute neutrophil
count (ANC) at the time of controlled CCI which correlated
with BBB damage in the injured hemisphere 24 hours later.
However, no difference in brain edema and hemispheric
neutrophil accumulation were reported between control and
G-CSF treated rats. They have suggested
that the ability of
G-CSF-stimulated neutrophils to migrate
into injured tissue
may be impaired (69). Recently, Park et al. have reported
that intra-peritoneal G-CSF administration after
intracerebral hemorrhage reduces brain edema and
inflammation in a rat model (17). However, they also
Neuroprotection by G-CSF
716
Table 1. Previous experimental studies about neuroprotective effect of G-CSF treatment after CNS injuries
Species Model Results Author Year Ref.
Rat
Pretreatment
TBI
No increase in brain edema and posttraumatic brain neutrophil accumulation;
Increase in BBB damage;
Whalen M. J., et al. 2000 69
Mice EAE
Limitation of demyelination and inflammation;
Clinical score improvement;
Reduction of pro-inflammatory cytokines;
Zavala F., et al. 2002 74
Mice MCAO
Improving survival rate;
Reduction of infarct volume;
Six I., et al. 2003 18
Rat
MCAO
Cell Culture
Reduction of infarct volume;
Reduction of inflammation;
Protection against excitotoxicity;
Schabitz W. R., et
al.
2003 20
Mice TBI
Small functional effect on functional outcome;
No effect on histopathology or motor outcome;
Sheibani N., et al. 2004 19
Rat MCAO
Neurological improvement;
Reduction of infarct volume;
Improving neural plasticity and vascularization;
Shyu W. C., et al. 2004 16
Rat ICH
Neurological improvement;
Reduction of edema, inflammation and
perihematomal cell death;
Park H. K., et al. 2005 17
Mice MCAO
Neurological improvement;
Reduction of infarct volume;
Gibson C. L., et al. 2005 11
Rat
MCAO
CCCA/MCAO
Cell culture
Neurological improvement;
Reduction of infarct volume;
Induce neurogenesis;
Schneider A., et al. 2005 12
TBI; traumatic brain injury, EAE; experimental allergic encephalomyelitis, MCAo; middle cerebral artery occlusion,
ICH; intracerebral hemorrhage, CCCA/MCAo; combined common carotid artery/ middle cerebral artery occlusion, BBB; blood
brain barier, Ref: Reference
showed that G-CSF treatment also decreases the BBB
permeability after injury leading to contradictory results
(17). The neuroprotective effect of G-CSF in neurons was
suggested by Schabitz et al., by demonstrating that intra-
venous administration of G-CSF after focal cerebral
ischemia reduced the volume of infarction and the number
of infiltrated neutrophils into the ischemic hemisphere 24
hours later. Furthermore, neuroprotective effect of G-CSF
is further supported by the finding that G-CSF protects
cerebellar granule cells exposed to glutamate in vitro (20).
Peripherally derived leukocytes not only release
proteolytic enzymes and free oxygen radicals but also
produce and secrete cytokines including tumor necrosis-
alpha (TNF-alpha) and interleukin-1-beta (IL-1beta) which
mediate BBB injury and enhance the migration of blood
leucocytes into the injured tissue by modulating the
expressions of various surface antigens (62, 70, 71). The
possible mechanisms of G-CSF treatment on the prevention
of BBB damage may be through its ability to decrease the
levels of proinflammatory cytokines. A
series of reports
have demonstrated that G-CSF displays immunoregulatory
properties through interaction with its receptor. It has been
shown that G-CSF treatment
protects rodents against
lipopolysaccharide (LPS)-induced lethal
toxicity by
suppressing systemic TNF-alpha
production in vivo (72).
Such reduced cytokine production as well as by an increase
in production of anti-inflammatory counterregulatory
molecules has also been shown in peripheral blood
leukocytes from G-CSF-treated
healthy volunteers (73).
Also, G-CSF reduces the T cell infiltration and
inflammation within the CNS in experimental
allergic
encephalomyelitis by reducing interferon-gamma (IFN-
gamma) and increasing IL-4 and TGF-beta1 levels, and
with a reduction of systemic and lymphocyte TNF-alpha
production (74). Nishiki et al. have suggested that G-CSF
inhibits LPS-induced TNF-alpha
production in human
monocytes
through selective activation of JAK2/STAT3
pathways (75).
G-CSF activates JAK2 in turn activates a
transcription
factor, STAT3, triggering a signaling
transduction cascade that leads
to increase in cellular
inhibitor of apoptosis protein2 (cIAP2) expression in
human neutrophils resulting in their survival (76) (Figure
1). Activation of STAT3 by G-CSF has also been linked to
myeloid cell differentiation
(77). However, STAT5 seems
to be involved in G-CSF-dependent cell
proliferation (78).
Also, G-CSF activation of JAK2-STAT3 pathway and the
resulting induction of antiapoptotic proteins and inhibition
of apoptotic death of cardiomyocytes in the infarcted hearts
have been recently reported by Harada et al. (79). Schabitz
et al. showed for the first time that G-CSF treatment results
in increased STAT3 expression in the penumbra of the
infarction after focal cerebral ischemia in rats and they
have suggested that upregulation of STAT3 in neurons may
mediate the antiapoptotic effects of G-CSF (20).
Antiapoptotic effect of G-CSF in neural tissue was also
pointed by Park et al. by demonstrating reduced number of
TUNEL positive neurons in the penumbra following G-
CSF treatment after intracerebral hemorrhage in rats (17).
More recently, G-CSF has been reported to
protect cortical neurons in vitro against camptothecin-
induced and NO-induced apoptosis by reducing caspase-3
and poly(ADP-ribose) polymerase (PARP) cleavage (12).
Authors showed that G-CSF exposure results in rapid
STAT3 phosphorylation by JAK2 kinase in neurons, which
was inhibited by AG 490, a specific inhibitor of JAK2. The
authors further reported that G-CSF leads to increase of
protein levels of the antiapoptotic STAT3 target Bcl-X
L
(Figure 2)
(12). However, determination of activation levels
of the extracellular signal-regulated kinase (ERK) family
showed that while ERK1/2 was transiently and weakly
Neuroprotection by G-CSF
717
Figure 2. Figure shows the signaling pathways implicated
in G-CSF mediated neuronal survival. Activation of the
extracellular signal-regulated kinase (ERK) family
enhances neuronal survival. PI3-K/Akt and STAT3
signaling
pathways when activated prevent apoptotic cell
death by inhibiting activation of caspases and increasing
antiapoptotic protein members such as Bcl-xL.
ctivated by G-CSF, ERK5 kinase was strongly activated in
cultured neurons from rat cortex (12). ERK5, also known
as big MAP kinase 1 (BMK1), is a MAPK member whose
biological role is largely
undefined. However, it has been
suggested that ERK5 activation enhances neuronal survival
(80, 81). Schneider et al. also showed the activation of PI3-
K/Phosphoinositide-dependent
kinase (PDK)/Akt signaling
pathway by G-CSF in cortical neurons in vitro. They
suggested that anti-apoptotic action of G-CSF on neurons
mediated at least partially by the PI3-K/Akt pathway (12)
(Figure 2). It is well established that Akt, a downstream
target of PI3-K, is a critical anti-apoptotic factor in
controlling the balance between survival and apoptosis in
multiple cell systems including neurons by several
mechanisms such as by phosphorylating Bcl-2-associated
death protein (BAD) and caspase-9, and inducing Bcl-2
expression (82-86). PDK has also been shown to
phosphorylate and activate Akt to promote cell survival
(87).
There is strong evidence from previous studies
that G-CSF exerts antiapoptotic effect on many
hematopoietic cell lines by triggering various signal
pathways. G-CSF inhibits spontaneous cytochrome c
release and mitochondria dependent apoptosis of
myelodysplastic syndrome hematopoietic progenitors (88).
It has been reported that G-CSF-induced survival of
neutrophils
associates with inhibition of cleavage of Bid,
Bid/Bax translocation
to the mitochondria, and prevention
of subsequent release of
the proapoptotic mitochondrial
constituents. Moreover, G-CSF blocks both processing of
the initiator
caspase-8 and caspase-9 and the executioner
caspase-3 and their
specific enzymatic activities in
apoptotic neutrophils (89). Furthermore, G-CSF inhibits the
processing of proIL-1beta and the release of mature IL-
1beta in LPS-stimulated whole blood via the inhibition of
caspase-1 which is also known as interleukin-converting
enzyme (90). It has also been shown that mice deficient in
interleukin-1 converting enzyme are resistant to neonatal
hypoxic-ischemic brain damage (91). These mechanisms
suggest probable mechanisms for the anti-apoptotic effects
of G-CSF treatment on hematopoietic cell lines. However,
it needs to be studied whether G-CSF maintains viability of
neuronal cells by similar mechanisms. The precise
mechanism by which G-CSF exerts anti-apoptotic function
in the neural tissue in vivo is not well understood.
5.2. G-CSF, hematopoietic stem cell mobilization, and
brain repair
There is a growing interest to restore brain
function after stroke by transplanting stem cells. Various
stem cells especially porcine fetal cells, neural stem cells
(NSCs), and bone marrow stem cells (BMCs) currently are
under investigation by various independent research groups
(92-95).
Recent studies have focused on the potential
plasticity of bone marrow stem cells (BMCs) after stroke.
There are two populations of bone marrow stem cells:
hematopoietic stem cells (HSCs), which can differentiate
into every type of mature blood cells; and mesenchymal
stem cells (MSCs), which can differentiate into adipose,
muscle, bone, cartilage, endothelium, hepatocyte, glia, and
neuron under appropriate stimuli in vitro and in vivo (96-
104). However, some controversy still exists about
differentiation of BMCs into neural tissues (105-107).
In previous studies, it has been reported that after
direct intrastriatal, intracarotid, and intravenous delivery,
MSCs migrate to the ischemic area, survive and
differentiate into neuronal and glial cell types, and improve
functional recovery after experimental stroke (92-94). The
migration of MSCs is supported by evidence that Y
chromosome-positive
neuron-like cells are seen in
postmortem brain samples from females who
had received
bone marrow transplants from male
donors in humans (95).
Although various studies attributed the beneficial
results of BMC treatment after stroke to possible neuronal
plasticity, the precise mechanisms of the functional benefits
still remains unclear. Several studies have showed
functional benefits may result either from enhanced
neurogenesis or from a favorable impact of stem cells or
their released cytokines which stimulate angiogenesis and
reduce cell death. It has been shown that treatment of
stroke with human MSCs enhances angiogenesis in the
host
brain which is mediated by increase
in levels of endogenous
rat vascular endothelial growth factor (VEGF) and VEGF-
Neuroprotection by G-CSF
718
receptor 2 (108). Other studies have shown reduction of
apoptosis, increase in BDNF and nerve growth factor in the
penumbral zone of the lesion, and the proliferation of
endogenous cells in the subventricular zone after
administration of human MSCs in rats (109). Moreover, it
has been suggested that intrastriatal transplantation of
mouse MSCs restores cerebral blood flow and BBB to
normal levels, elevates expression levels of neurotrophic
factors including activin A, the glial cell line-derived
neurotrophic factor, and transforming growth factor-beta 1
and 2 (110). So, these mechanisms of recovery may also be
due to the release of trophic factors into the damaged area
rather than neuronal differentiation and implant integration
to the injured ischemic site.
Recently, Bang et al. have examined the
feasibility, efficacy, and safety of cell therapy using
culture-expanded autologous MSCs in patients with
ischemic stroke (111). They have reported that intravenous
infusion of 1x10
8
autologous MSCs to patients, more than 1
month after the onset of stroke symptoms, improves
outcome (111). However, they stress the need of future
double-blind studies with larger cohorts to reach a
definitive conclusion regarding the efficacy of MSC
therapy (111).
It is well known that administration of G-CSF
mobilizes HSCs from bone
marrow into peripheral blood
(13). G-CSF has been shown to result in significant
decrease in infarct volume and enhance survival rate after
focal cerebral ischemia in mice, and these benefits have
been suggested to be mediated by mobilization of
autologous HSCs into circulation and their contribution to
brain repair (18). Moreover, Shyu et al. showed that G-CSF
treatment increases BrdU
+
cells coexpressing the neuronal
phenotypes of Neu-N
+
and microtubule-associated protein-
2 (MAP-2)
+
cells as well as the glial phenotype of GFAP
+
cells in the ischemic cortical areas of G-CSF-treated rats
after focal cerebral ischemia in rats (16). They have
suggested that G-CSF treatment enhances translocation of
HSCs into ischemic brain, and significantly improves
lesion repair by improving neuronal plasticity and
vascularization (16). Interestingly, they have reported that
neutralization of CXC chemokine receptor 4 (CXCR4) by
its specific antibody results in a slight reduction in infarct
volume in G-CSF-treated rats (16).
The chemokine receptor CXCR4 is expressed in
cells of both the immune and the central nervous systems
and can mediate migration of resting leukocytes and
hematopoietic progenitors in response to its ligand, stromal
cell-derived factor-1 (SDF-1). SDF-1, the strong
chemoattractant was also shown
to be crucial for cerebellar
development (112, 113). Moreover, it has been suggested
that SDF-1/CXCR4 system may control cerebral infiltration
of CXCR4-carrying leukocytes
during cerebral ischemia,
and
contribute to ischemia-induced neuronal plasticity
(114). SDF-1 also play pivotal role in the
migration of
NSCs toward the ischemic core and penumbra, and
enhances proliferation,
promotes chain migration and
activates intracellular
molecular pathways mediating
engagement (115). Both microglial cells and astrocytes in
humans can express chemokine receptors including CCR4,
CCR5, CCR6, CXCR2, and CXCR4 and activation of the
cells with TNF-alpha and IFN-gamma changes the
expression levels of these chemokine receptors (116, 117).
CXCR4-mediated signaling in astroglioma
cells also
provides pathway for these cells to express chemokines
involved in angiogenesis and inflammation (118). Taken
together these data, a better understanding
of this complex
dynamic and its relationships with G-CSF may permit us to
understand precise mechanisms of G-CSF related
functional recovery after stroke.
5.3. G-CSF and neurogenesis
The subgranular zone of dentate gyrus (DG) in
the hippocampus and subventricular zone (SVZ) of the
lateral ventricle are the main areas of the adult brain that
undergo neurogenesis and gliogenesis (119-121). The SVZ
in the forebrain is the largest source of neural stem cells
(NSCs) and progenitor cells which can differentiate into
neurons, astrocytes, and oligodendrocytes (120, 122). There
is accumulating evidence indicating that this
neurogenerative capacity of the adult CNS could open
novel therapeutic approaches to neurodegenerative diseases
based on the use of NSC transplantation. Previous in vivo
experiments showed that transplanted human NSCs
survive, migrate, and differentiate in host brain and
promote functional recovery after intracerebral hemorrhage
and cerebral ischemia (123-125). Endogenous NSCs
sustain neurogenesis and gliogenesis in response to several
different injuries including ischemic and traumatic brain
injury (126-128).
It has been reported that outcome from ischemia
appears to be worse when neurogenesis is inhibited by
irradiation in gerbils (129). However, augmentation of this
self-repair phenomenon by exogenous agents can further
enhance neurogenesis and
might also have therapeutic
applications after CNS disorders. Enhanced neurogenesis
with growth factors such as EPO or heparin-binding
epidermal growth factor-like growth factor (HB-EGF) have
been reported to decrease infarct size and improve
neurological functions after stroke in rats (130, 131).
More recently, G-CSF has been shown to have
functional role in differentiation of adult rat NSCs both in
vitro and in vivo (12). Analysis of G-CSF responsiveness in
the cultured NSCs from the rat SVZ or hippocampal region
indicates that G-CSF exposure results mainly in increase in
the population of cells expressing mature neural markers
such as beta-III-tubulin, neuron-specific enolase (NSE),
and MAP-2 without elevating the number of immature stem
cells (12). Schneider et al. found that peripheral infusion of
G-CSF enhances recruitment of progenitor cells from the
lateral ventricular wall into the ischemic area of the
neocortex in rat (12). Moreover, authors reported that G-
CSF increases hippocampal neurogenesis not only in
ischemic animals, but also in the intact, non-ischemic rat.
Based on this evidence, they argued that G-CSF may
enhance structural repair and function even in healthy
subjects or at long intervals after stroke (12).
Neuroprotection by G-CSF
719
The intracellular mechanisms that regulate
neurogenesis both in normal and ischemic conditions
remain unclear. Increased cyclic guanosine monophosphate
(cGMP), a molecular messenger involved in regulation of
cellular proliferation, has been reported to enhance
neurogenesis (132). Recently, Wang et al. showed that PI3-
K/Akt pathway mediates cGMP enhanced proliferation of
adult progenitor cells derived from the SVZ of the rat
(133). Moreover, it has been suggested that stroke-
enhanced neuroblast migration is independent of cell
proliferation and survival, and PI3-K/Akt signal
transduction pathway mediates neuroblast migration after
stroke (134). The activation of PI3-K/Akt signaling
pathway by G-CSF has been shown in cortical neurons in
vitro (12). The anti-apoptotic effect of G-CSF on neurons is
dependent on activation of PI3-K/Akt signaling
pathway. It
can be further hypothesized that G-CSF may also augment
neurogenesis and neuroblast migration by activation of PI3-
K/Akt signaling
pathway.
It has been shown that CXCR4 is expressed in rat
and human neural progenitor cells (135). The
CXCR4/SDF-1 system is important in mediating specific
migration of neural progenitor cells to the site of ischemic
damaged neurons (115, 136). As discussed previously,
neutralization of CXCR4 by its specific antibody has been
reported to reduce neuroprotective effect of G-CSF in rats
after stroke (16). Thus, it can be hypothesized that
CXCR4/SDF-1 system may play important role in G-CSF
induced neurogenesis. Additional studies however will be
necessary to clarify this issue.
It is well known that the regulation of adult
neurogenesis and the maintenance of stem cell renewal are
modulated by various regulatory mechanisms including
growth factors. Although the ability of G-CSF to stimulate
the neurogenesis shows a lot of promise, additional studies
are required for determining the molecular basis of this
effect.
6. SUMMARY AND PERSPECTIVE
An increasing amount of data suggests a
neuroprotective role for G-CSF. The molecular
mechanisms of neuroprotective effect of G-CSF especially
under in vivo settings still remain unclear. However, the
neuroprotective property of G-CSF may be due, at least in
part, to the mobilization of BMCs to the injured brain.
However, the precise mechanism of BMCs-mediated brain
repair and/or neuroprotection after stroke is still under
investigation. Furthermore, G-CSF has anti-apoptotic, anti-
inflammatory, immunomodulatory, and neurogenesis-
inducing effects which provide other potential mechanisms
that may be responsible for neuroprotection and long term
benefits.
Neuroprotective effects of G-CSF however,
should be tested in multiple animal models in different
species with different time points, different doses and
application routes to confirm results. Also, the use of G-
CSF in conjunction with rt-PA should be tested for safety
before starting clinical trials in humans.
7. ACKNOWLEDGEMENT
This study was partially supported by grants from
NIH NS45694, HD43120, and NS43338 to JHZ.
8. REFERENCES
1. Heart Disease and Stroke Statistics -2005 Update,
American Heart Association
2. The National Institute of Neurological Disorders and
Stroke rt-PA Stroke Study Group. Tissue plasminogen
activator for acute ischemic stroke. N Engl J Med 333(24),
1581-1587 (1995)
3. Wang Y. F., S. E. Tsirka, S. Strickland, P. E. Stieg, S. G.
Soriano & S. A. Lipton: Tissue plasminogen activator
(tPA) increases neuronal damage after focal cerebral
ischemia in wild-type and tPA-deficient mice. Nat Med
4(2), 228-231 (1998)
4. Tsirka S. E., A. Gualandris, D. G. Amaral & S.
Strickland: Excitotoxin-induced neuronal degeneration and
seizure are mediated by tissue plasminogen activator.
Nature 377(6547), 340-344 (1995)
5. Sun Y., J. W. Calvert & J. H. Zhang: Neonatal
hypoxia/ischemia is associated with decreased
inflammatory mediators after erythropoietin administration.
Stroke 36(8), 1672-1678 (2005)
6. Solaroglu I., A. Solaroglu, E. Kaptanoglu, S. Dede, A.
Haberal, E. Beskonakli & K. Kilinc: Erythropoietin
prevents ischemia-reperfusion from inducing oxidative
damage in fetal rat brain. Childs Nerv Syst 19(1), 19-22
(2003)
7. Kaptanoglu E., I. Solaroglu, O. Okutan, H. S. Surucu, F.
Akbiyik & E. Beskonakli: Erythropoietin exerts
neuroprotection after acute spinal cord injury in rats: effect
on lipid peroxidation and early ultrastructural findings.
Neurosurg Rev 27(2), 113-120 (2004)
8. Sun Y, C. Zhou, P. Polk, A. Nanda & J. H. Zhang:
Mechanisms of erythropoietin-induced brain protection in
neonatal hypoxia-ischemia rat model. J Cereb Blood Flow
Metab 24(2), 259-270 (2004)
9. Schabitz W. R., T. T. Hoffmann, S. Heiland, R. Kollmar,
J. Bardutzky, C. Sommer & S. Schwab: Delayed
neuroprotective effect of insulin-like growth factor-I after
experimental transient focal cerebral ischemia monitored
with MRI. Stroke 32(5), 1226-1233 (2001)
10. Schabitz W. R., C. Sommer, W. Zoder, M. Kiessling,
M. Schwaninger & S. Schwab: Intravenous brain-derived
neurotrophic factor reduces infarct size and
counterregulates Bax and Bcl-2 expression after temporary
focal cerebral ischemia. Stroke 31(9), 2212-2217 (2000)
11. Gibson C. L., P. M. Bath & S. P. Murphy: G-CSF
reduces infarct volume and improves functional outcome
after transient focal cerebral ischemia in mice. J Cereb
Blood Flow Metab 25(4), 431-439 (2005)
12. Schneider A.,C. Kruger, T. Steigleder, D. Weber,
C.
Pitzer, R. Laage,
J. Aronowski, M. H. Maurer, N. Gassler,
W. Mier, M. Hasselblatt, R. Kollmar, S. Schwab,
C.
Sommer, A. Bach,
H. G. Kuhn & A. W. Schabitz: The
hematopoietic factor G-CSF is a neuronal ligand that
counteracts programmed cell death and drives
neurogenesis. J Clin Invest 115(8), 2083-2098 (2005)
Neuroprotection by G-CSF
720
13. Demetri G. D. & J. D. Griffin: Granulocyte colony-
stimulating factor and its receptor. Blood 78(11), 2791-
2808 (1991)
14. Konishi Y., D. H. Chui, H. Hirose, T. Kunishita & T.
Tabira: Trophic effects of erythropoietin and other
hematopoietic factors on central cholinergic neurons in
vitro and in vivo. Brain Res 609(1-2), 29-35 (1993)
15. Lin L. F., D. H. Doherty, J. D. Lile, S. Bektesh & F.
Collins: GDNF: a glial cell line-derived neurotrophic factor
for midbrain dopaminergic neurons. Science 260(5111),
1130-1132 (1993)
16. Shyu W. C., S. Z. Lin, H. I. Yang, Y. S. Tzeng, C. Y.
Pang, P. S. Yen & H. Li: Functional recovery of stroke rats
induced by granulocyte colony-stimulating factor-
stimulated stem cells. Circulation 110(13), 1847-1854
(2004)
17. Park H. K., K. Chu, S. T. Lee, K. H. Jung, E. H. Kim,
K. B. Lee, Y. M. Song, S. W. Jeong, M. Kim & J. K. Roh:
Granulocyte colony-stimulating factor induces
sensorimotor recovery in intracerebral hemorrhage. Brain
Res 1041(2), 125-131 (2005)
18. Six I., G. Gasan, E. Mura & R. Bordet: Beneficial effect
of pharmacological mobilization of bone marrow in
experimental cerebral ischemia. Eur J Pharmacol 458(3),
327-328 (2003)
19. Sheibani N., E. F. Grabowski, D. A. Schoenfeld, M. J.
Whalen: Effect of granulocyte colony-stimulating factor on
functional and histopathologic outcome after traumatic
brain injury in mice. Crit Care Med 32(11), 2274-2278
(2004)
20. Schabitz W. R., R. Kollmar, M. Schwaninger, E.
Juettler, J. Bardutzky, M. N. Scholzke, C. Sommer & S.
Schwab: Neuroprotective effect of granulocyte colony-
stimulating factor after focal cerebral ischemia. Stroke
34(3), 745-751 (2003)
21. Wells J. A. & A. M. de Vos: Hematopoietic receptor
complexes. Annu Rev Biochem 65, 609-634 (1996)
22. Aloisi F., A. Care, G. Borsellino, P. Gallo, S. Rosa, A.
Bassani, A. Cabibbo, U. Testa, G. Levi & C. Peschle:
Production of hemolymphopoietic cytokines (IL-6, IL-8,
colony-stimulating factors) by normal human astrocytes in
response to IL-1 beta and tumor necrosis factor-alpha. J
Immunol 149(7), 2358-2366 (1992)
23. Malipiero U. V., K. Frei & A. Fontana: Production of
hemopoietic colony-stimulating factors by astrocytes. J
Immunol 144(10):3816-3821 (1990)
24. Le Beau M., R. Lemons, J. Carrino, M. Pettenati, L. Souza,
M. Diaz & J. Rowley: Chromosomal localization of the human
G-CSF gene to 17q11 proximal to the breakpoint of the
t(15;17) in acute promyelocytic leukemia. Leukemia 1(12),
795-799 (1987)
25. Simmers R. N., L. M. Webber, M. F. Shannon, O. M.
Garson, G. Wong, M. A. Vadas & G. R. Sutherland:
Localization of the G-CSF gene on chromosome 17 proximal
to the breakpoint in the t(15;17) in acute promyelocytic
leukemia. Blood 70(1), 330-332 (1987)
26. Fukunaga R., E. Ishizaka-Ikeda, C. X. Pan, Y. Seto & S.
Nagata: Functional domains of the granulocyte colony-
stimulating factor receptor. EMBO J 10(10), 2855-2865 (1991)
27. Budel L. M., I. P. Touw, R. Delwel & B. Lowenberg:
Granulocyte colony-stimulating factor receptors in human
acute myelocytic leukemia. Blood 74(8), 2668-2673 (1989)
28. Hanazono Y., T. Hosoi, T. Kuwaki, S. Matsuki, K.
Miyazono, K. Miyagawa & F. Takaku: Structural analysis
of the receptors for granulocyte colony-stimulating factor
on neutrophils. Exp Hematol 18(10), 1097-1103 (1990)
29. Shimoda K., S. Okamura, N. Harada, S. Kondo, T.
Okamura & Y. Niho: Identification of a functional receptor
for granulocyte colony-stimulating factor on platelets. J
Clin Invest 91(4), 1310-1313 (1993)
30. Morikawa K., S. Morikawa, M. Nakamura & T.
Miyawaki: Characterization of granulocyte colony-
stimulating factor receptor expressed on human
lymphocytes. Br J Haematol 118(1), 296-304 (2002)
31. Boneberg E. M., L. Hareng, F. Gantner, A. Wendel &
T. Hartung: Human monocytes express functional receptors
for granulocyte colony-stimulating factor that mediate
suppression of monokines and interferon-gamma. Blood
95(1), 270-276 (2000)
32. Bussolino F., J. M. Wang, P. Defilippi, F. Turrini, F.
Sanavio, C. J. Edgell, M. Aglietta, P. Arese & A.
Mantovani: Granulocyte- and granulocyte-macrophage-
colony stimulating factors induce human endothelial cells
to migrate and proliferate. Nature 337(6206):471-473
(1989)
33. Shimoda K., J. Feng, H. Murakami, S. Nagata, D.
Watling, N. C. Rogers, G. R. Stark, I. M. Kerr & J. N. Ihle:
Jak1 plays an essential role for receptor phosphorylation
and Stat activation in response to granulocyte colony-
stimulating factor. Blood 90(2), 597-604 (1997)
34. Tian S. S., P. Lamb, H. M. Seidel, R. B. Stein & J.
Rosen: Rapid activation of the STAT3 transcription factor
by granulocyte colony-stimulating factor. Blood 84(6),
1760-1764 (1994)
35. Hunter M. G. & B. R. Avalos: Phosphatidylinositol 3'-
kinase and SH2-containing inositol phosphatase (SHIP) are
recruited by distinct positive and negative growth-
regulatory domains in the granulocyte colony-stimulating
factor receptor. J Immunol 160(10), 4979-4987 (1998)
36. Dong F. & A. C. Larner: Activation of Akt kinase by
granulocyte colony-stimulating factor (G-CSF): evidence
for the role of a tyrosine kinase activity distinct from the
Janus kinases. Blood 95(5), 1656-1662 (2000)
37. Ward A. C., D. M. Loeb, A. A. Soede-Bobok, I. P.
Touw & A. D. Friedman: Regulation of granulopoiesis by
transcription factors and cytokine signals. Leukemia 14(6),
973-990 (2000)
38. Kleinschnitz C., M. Schroeter, S. Jander & G. Stoll:
Induction of granulocyte colony-stimulating factor mRNA by
focal cerebral ischemia and cortical spreading depression.
Brain Res Mol Brain Res 131(1-2),73-78 (2004)
39. Grasso G., A. Sfacteria, A. Cerami & M. Brines:
Erythropoietin as a tissue-protective cytokine in brain injury:
what do we know and where do we go? Neuroscientist 10(2),
93-98 (2004)
40. Buemi M., E. Cavallaro, F. Floccari, A. Sturiale, C. Aloisi,
M. Trimarchi, F. Corica & N. Frisina: The pleiotropic effects
of erythropoietin in the central nervous system. J Neuropathol
Exp Neurol 62(3), 228-236 (2003)
41. Welte K., J. Gabrilove, M. H. Bronchud, E. Platzer & G.
Morstyn: Filgrastim (r-metHuG-CSF): the first ten years.
Blood 88(6), 1907-1929 (1996)
42. Carlsson G., A. Ahlin, G. Dahllof, G. Elinder, J. I.
Henter & J. Palmblad: Efficacy and safety of two different
Neuroprotection by G-CSF
721
rG-CSF preparations in the treatment of patients with
severe congenital neutropenia. Br J Haematol 126(1), 127-
132 (2004)
43. Bensinger W. I., C. H. Weaver, F. R. Appelbaum, S.
Rowley, T. Demirer, J. Sanders, R. Storb & C. D. Buckner:
Transplantation of allogeneic peripheral blood stem cells
mobilized by recombinant human granulocyte colony-
stimulating factor. Blood 85(6), 1655-1658 (1995)
44. Kocherlakota P. & E. F. La Gamma: Human
granulocyte colony-stimulating factor may improve
outcome attributable to neonatal sepsis complicated by
neutropenia. Pediatrics 100(1), E6 (1997)
45. Wunderink R., Jr. K. Leeper, R. Schein, S. Nelson, B.
DeBoisblanc, N. Fotheringham & E. Logan: Filgrastim in
patients with pneumonia and severe sepsis or septic shock.
Chest 119(2), 523-529 (2001)
46. Gough A., M. Clapperton, N. Rolando, A. V. Foster, J.
Philpott-Howard & M. E. Edmonds: Randomised placebo-
controlled trial of granulocyte-colony stimulating factor in
diabetic foot infection. Lancet 350(9081), 855-859 (1997)
47. Nelson S., A. M. Heyder, J. Stone, M. G. Bergeron, S.
Daugherty, G. Peterson, N. Fotheringham, W. Welch, S.
Milwee & R. Root. A randomized controlled trial of
filgrastim for the treatment of hospitalized patients with
multilobar pneumonia. J Infect Dis 182(3), 970-973 (2000)
48. Schafer H., K. Hubel, H. Bohlen, G. Mansmann, K.
Hegener, B. Richarz, F. Oberhauser, G. Wassmer, A. H.
Holscher, H. Pichlmaier, V. Diehl & A. Engert:
Perioperative treatment with filgrastim stimulates
granulocyte function and reduces infectious complications
after esophagectomy. Ann Hematol 79(3), 143-151 (2000)
49. Schneider C., S. von Aulock, S. Zedler, C. Schinkel, T.
Hartung & E. Faist: Perioperative recombinant human
granulocyte colony-stimulating factor (Filgrastim)
treatment prevents immunoinflammatory dysfunction
associated with major surgery. Ann Surg 239(1), 75-81
(2004)
50. Mitsuyasu R.: Prevention of bacterial infections in
patients with advanced HIV infection. AIDS 13(Suppl 2),
S19-23 (1999)
51. Cruciani M., B. A. Lipsky, C. Mengoli & F. de Lalla:
Are granulocyte colony-stimulating factors beneficial in
treating diabetic foot infections?: A meta-analysis.
Diabetes Care 28(2), 454-460 (2005)
52. Schaefer H., A. Engert, G. Grass, G. Mansmann, G.
Wassmer, K. Hubel, D. Loehlein, B. C. Ulrich, H. Lippert,
W. T. Knoefel & A. H. Hoelscher: Perioperative
granulocyte colony-stimulating factor does not prevent
severe infections in patients undergoing esophagectomy for
esophageal cancer: a randomized placebo-controlled
clinical trial. Ann Surg 240(1), 68-75 (2004)
53. Carr R., N. Modi & C. Dore: G-CSF and GM-CSF for
treating or preventing neonatal infections. Cochrane
Database Syst Rev (3), CD003066 (2003)
54. Cheng A. C., D. P. Stephens & B. J. Currie:
Granulocyte-Colony Stimulating Factor (G-CSF) as an
adjunct to antibiotics in the treatment of pneumonia in
adults. Cochrane Database Syst Rev (3), CD004400 (2004)
55. Heard S. O., M. P. Fink, R. L. Gamelli, J. S. Solomkin,
M. Joshi, A. L. Trask, T. C. Fabian, L. D. Hudson, K. B.
Gerold & E. D. Logan: Effect of prophylactic
administration of recombinant human granulocyte colony-
stimulating factor (filgrastim) on the frequency of
nosocomial infections in patients with acute traumatic brain
injury or cerebral hemorrhage. The Filgrastim Study
Group. Crit Care Med 26(4):748-754 (1998)
56. Whalen M. J., T. M. Carlos, R. S. Clark & P. M.
Kochanek: An acute inflammatory response to the use of
granulocyte colony-stimulating factor to prevent infections
in patients with brain injury: what about the brain? Crit
Care Med 27(5), 1014-1018 (1999)
57. Shenaq S. A., D. H. Yawn, A. Saleem, R. Joswiak & E.
S. Crawford: Effect of profound hypothermia on leukocytes
and platelets. Ann Clin Lab Sci 16(2), 130-133 (1986)
58. Bohn D. J., W. D. Biggar, C. R. Smith, A. W. Conn &
G. A. Barker: Influence of hypothermia, barbiturate
therapy, and intracranial pressure monitoring on morbidity
and mortality after near-drowning. Crit Care Med 14(6),
529-534 (1986)
59. Ishikawa K., H. Tanaka, M. Takaoka, H. Ogura, T.
Shiozaki, H. Hosotsubo, T. Shimazu, T. Yoshioka & H.
Sugimoto: Granulocyte colony-stimulating factor
ameliorates life-threatening infections after combined
therapy with barbiturates and mild hypothermia in patients
with severe head injuries. J Trauma 46(6), 999-1007
(1999)
60. Arand M., H. Melzner, L. Kinzl, U. B. Bruckner & F.
Gebhard: Early inflammatory mediator response following
isolated traumatic brain injury and other major trauma in
humans. Langenbecks Arch Surg 386(4), 241-248 (2001)
61. Dirnagl U., C. Iadecola & M. A. Moskowitz:
Pathobiology of ischaemic stroke: an integrated view.
Trends Neurosci 22(9), 391-397 (1999)
62. Barone F. C. & G. Z. Feuerstein: Inflammatory
mediators and stroke: new opportunities for novel
therapeutics. J Cereb Blood Flow Metab 19(8), 819-834
(1999)
63. Hayward N. J., P. J. Elliott, S. D. Sawyer, R. T.
Bronson & R. T. Bartus: Lack of evidence for neutrophil
participation during infarct formation following focal
cerebral ischemia in the rat. Exp Neurol 139(2), 188-202
(1996)
64. Freeman B. A. & J. D. Crapo: Free radicals and tissue
injury. Lab Invest 47 (5), 412-426 (1982)
65. Halliwell B. & O. I. Aruoma: DNA damage by oxygen-
derived species. FEBS Letter 281(1-2), 9-19 (1991)
66. Slater T. F.: Free-radical mechanisms in tissue injury.
Biochem J 222(1), 1-15 (1984)
67. Wei E. P., M. D. Ellison, H. A. Kontos & J. T.
Povlishock: O2 radicals in arachidonate-induced increased
blood-brain barrier permeability to proteins. Am J Physiol
251(4 Pt 2), H693-699 (1986)
68. Giulian D., J. Chen, J. E. Ingeman, J. K. George & M.
Noponen: The role of mononuclear phagocytes in wound
healing after traumatic injury to adult mammalian brain. J
Neurosci 9(12):4416-4429 (1989)
69. Whalen M. J., T. M. Carlos, S. R. Wisniewski, R. S.
Clark, J. A. Mellick, D. W. Marion & P. M. Kochanek:
Effect of neutropenia and granulocyte colony stimulating
factor-induced neutrophilia on blood-brain barrier
permeability and brain edema after traumatic brain injury in
rats. Crit Care Med 8(11), 3710-3717 (2000)
70. Quagliarello V. J., B. Wispelwey, W. J. Jr. Long & W.
M. Scheld: Recombinant human interleukin-1 induces
Neuroprotection by G-CSF
722
meningitis and blood-brain barrier injury in the rat.
Characterization and comparison with tumor necrosis
factor. J Clin Invest 87(4), 1360-1366 (1991)
71. Rosenman S. J., P. Shrikant, L. Dubb, E. N. Benveniste
& R. M. Ransohoff: Cytokine-induced expression of
vascular cell adhesion molecule-1 (VCAM-1) by astrocytes
and astrocytoma cell lines. J Immunol 154(4), 1888-1899
(1995)
72. Gorgen I., T. Hartung, M. Leist, M. Niehorster, G.
Tiegs, S. Uhlig, F. Weitzel & A. Wendel: Granulocyte
colony-stimulating factor treatment protects rodents against
lipopolysaccharide-induced toxicity via suppression of
systemic tumor necrosis factor-alpha. J Immunol 149(3),
918-924 (1992)
73. Hartung T., W. D. Docke, F. Gantner, G. Krieger, A.
Sauer, P. Stevens, H. D. Volk & A. Wendel: Effect of
granulocyte colony-stimulating factor treatment on ex vivo
blood cytokine response in human volunteers. Blood 85(9),
2482-2489 (1995)
74. Zavala F., S. Abad, S. Ezine, V. Taupin, A. Masson &
J. F. Bach: G-CSF therapy of ongoing experimental allergic
encephalomyelitis via chemokine- and cytokine-based
immune deviation. J Immunol 168(4), 2011-2019 (2002)
75. Nishiki S., F. Hato, N. Kamata, E. Sakamoto, T.
Hasegawa, A. Kimura-Eto, M. Hino & S. Kitagawa:
Selective activation of STAT3 in human monocytes
stimulated by G-CSF: implication in inhibition of LPS-
induced TNF-alpha production. Am J Physiol Cell Physiol
286(6), 1302-1311 (2004)
76. Hasegawa T., K. Suzuki, C. Sakamoto, K. Ohta, S.
Nishiki, M. Hino, N. Tatsumi & S. Kitagawa: Expression
of the inhibitor of apoptosis (IAP) family members in
human neutrophils: up-regulation of cIAP2 by granulocyte
colony-stimulating factor and overexpression of cIAP2 in
chronic neutrophilic leukemia. Blood 101(3), 1164-1171
(2003)
77. Shimozaki K., K. Nakajima, T. Hirano & S. Nagata:
Involvement of STAT3 in the granulocyte colony-
stimulating factor-induced differentiation of myeloid cells.
J Biol Chem 272(40), 25184-25189 (1997)
78. Dong F., X. Liu, J. P. de Koning, I. P. Touw, L.
Hennighausen, A. Larner & P. M. Grimley: Stimulation of
Stat5 by granulocyte colony-stimulating factor (G-CSF) is
modulated by two distinct cytoplasmic regions of the G-
CSF receptor. J Immunol 161(12), 6503-6509 (1998)
79. Harada M., Y. Qin, H. Takano, T. Minamino, Y. Zou,
H. Toko, M. Ohtsuka, K. Matsuura, M. Sano, J. Nishi, K.
Iwanaga, H. Akazawa, T. Kunieda, W. Zhu, H. Hasegawa,
K. Kunisada, T. Nagai, H. Nakaya, K. Yamauchi-Takihara
& I. Komuro: G-CSF prevents cardiac remodeling after
myocardial infarction by activating the Jak-Stat pathway in
cardiomyocytes. Nat Med 11(3), 305-311 (2005)
80. Liu L., J. E. Cavanaugh,
Y. Wang,
H. Sakagami, Z.
Mao & Z. Xia: ERK5 activation of MEF2-mediated gene
expression plays a critical role in BDNF-promoted survival
of developing but not mature cortical neurons. Proc Natl
Acad Sci USA 100(14), 8532–8537 (2003)
81. Watson F. L., H. M. Heerssen, A. Bhattacharyya, L.
Klesse, M. Z. Lin & R. A. Segal: Neurotrophins use the
Erk5 pathway to mediate a retrograde survival response.
Nat Neurosci 4(10), 981-988 (2001)
82. Fujio Y., K. Guo, T. Mano, Y. Mitsuuchi, J.R. Testa &
K. Walsh: Cell cycle withdrawal promotes myogenic
induction of Akt, a positive modulator of myocyte survival.
Mol Cell Biol 19(7), 5073-5082 (1999)
83. Dudek H., S. R. Datta, T. F. Franke, M. J. Birnbaum, R.
Yao, G. M. Cooper, R. A. Segal, D. R. Kaplan & M. E.
Greenberg: Regulation of neuronal survival by the serine-
threonine protein kinase Akt. Science 275(5300), 661-665
(1997)
84. Del Peso L., M. Gonzalez-Garcia, C. Page, R. Herrera
& G. Nunez: Interleukin-3-induced phosphorylation of
BAD through the protein kinase Akt. Science 278(5338),
687-689 (1997)
85. Pugazhenthi S., A. Nesterova, C. Sable, K. A.
Heidenreich, L. M. Boxer, L. E. Heasley & J. E. Reusch:
Akt/protein kinase B up-regulates Bcl-2 expression through
cAMP-response element-binding protein. J Biol Chem
275(15), 10761-10766 (2000)
86. Cardone M. H., N. Roy, H. R. Stennicke, G. S.
Salvesen, T. F. Franke, E. Stanbridge, S. Frisch & J. C.
Reed: Regulation of cell death protease caspase-9 by
phosphorylation. Science 282(5392), 1318-1321 (1998)
87. Kim S., K. Jee, D. Kim, H. Koh & J. Chung: Cyclic
AMP inhibits Akt activity by blocking the membrane
localization of PDK1. J Biol Chem 276(16), 12864-12870
(2001)
88. Tehranchi R., B. Fadeel, A. M. Forsblom, B.
Christensson, J. Samuelsson, B. Zhivotovsky & E.
Hellstrom-Lindberg: Granulocyte colony-stimulating factor
inhibits spontaneous cytochrome-c release and
mitochondria-dependent apoptosis of myelodysplastic
syndrome hematopoietic progenitors. Blood 101(3), 1080-
1086 (2003)
89. Maianski N. A., D. Roos & T. W. Kuijpers: Bid
truncation, Bid/Bax targeting to the mitochondria, and
caspase activation associated with neutrophil apoptosis are
inhibited by granulocyte colony-stimulating factor. J
Immunol 172(11), 7024-7030 (2004)
90. Boneberg E. M. & T. Hartung: Granulocyte colony-
stimulating factor attenuates LPS-stimulated IL-1beta
release via suppressed processing of proIL-1beta, whereas
TNF-α release is inhibited on the level of proTNF-α
formation. Eur J Immunol 32(6), 1717-1725 (2002)
91. Liu X. H., D. Kwon, G. P. Schielke, G. Y. Yang, F. S.
Silverstein & J. D. Barks: Mice deficient in interleukin-1
converting enzyme are resistant to neonatal hypoxic-
ischemic brain damage. J Cereb Blood Flow Metab 19(10),
1099-1108 (1999)
92. Li Y., M. Chopp, J. Chen, L. Wang, S. C. Gautam, Y.
X. Xu & Z. Zhang: Intrastriatal transplantation of bone
marrow nonhematopoietic cells improves functional
recovery after stroke in adult mice. J Cereb Blood Flow
Metab 20(9), 1311-1319 (2000)
93. Li Y., J. Chen, L. Wang, M. Lu & M. Chopp:
Treatment of stroke in rat with intracarotid administration
of marrow stromal cells. Neurology 56(12), 1666-1672
(2001)
94. Chen J., Y. Li, L. Wang, Z. Zhang, D. Lu, M. Lu & M.
Chopp: Therapeutic benefit of intravenous administration
of bone marrow stromal cells after cerebral ischemia in
rats. Stroke 32(4), 1005-1011 (2001)
Neuroprotection by G-CSF
723
95. Mezey E., S. Key, G. Vogelsang, I. Szalayova, G. D.
Lange & B. Crain: Transplanted bone marrow generates
new neurons in human brains. Proc Natl Acad Sci USA
100(3), 1364-1369 (2003)
96. Grove J. E., E. Bruscia & D. S. Krause: Plasticity of
bone marrow-derived stem cells. Stem Cells 22(4), 487-
500 (2004)
97. Herzog E. L., L. Chai & D. S. Krause: Plasticity of
marrow-derived stem cells. Blood 102(10), 3483-3493
(2003)
98. Pittenger M. F., A. M. Mackay, S. C. Beck, R. K.
Jaiswal, R. Douglas, J. D. Mosca, M. A. Moorman, D. W.
Simonetti, S. Craig & D. R. Marshak: Multilineage
potential of adult human mesenchymal stem cells. Science
284(5411), 143-147 (1999)
99. Prockop D. J.: Marrow stromal cells as stem cells for
nonhematopoietic tissues. Science 276(5309), 71-74 (1997)
100. Petersen B. E., W. C. Bowen, K. D. Patrene, W. M.
Mars, A. K. Sullivan, N. Murase, S. S. Boggs, J. S.
Greenberger & J. P. Goff: Bone marrow as a potential
source of hepatic oval cells. Science 284(5417), 1168-1170
(1999)
101. Sanchez-Ramos J., S. Song, F. Cardozo-Pelaez, C.
Hazzi, T. Stedeford, A. Willing, T. B. Freeman, S. Saporta,
W. Janssen, N. Patel, D. R. Cooper & P. R. Sanberg: Adult
bone marrow stromal cells differentiate into neural cells in
vitro. Exp Neurol 164(2), 247-256 (2000)
102. Eglitis M. A. & E. Mezey: Hematopoietic cells
differentiate into both microglia and macroglia in the brains
of adult mice. Proc Natl Acad Sci USA 94(8), 4080-4085
(1997)
103. Hess D. C., W. D. Hill, A. Martin-Studdard, J. Carroll,
J. Brailer & J. Carothers: Bone marrow as a source of
endothelial cells and NeuN-expressing cells after stroke.
Stroke 33(5), 1362-1368 (2002)
104. Woodbury D., E. J. Schwarz, D. J. Prockop & I. B.
Black: Adult rat and human bone marrow stromal cells
differentiate into neurons. J Neurosci Res 61(4), 364-370
(2000)
105. Wehner T., M. Bontert, I. Eyupoglu, K. Prass, M.
Prinz, F. F. Klett, M. Heinze, I. Bechmann, R. Nitsch, F.
Kirchhoff, H. Kettenmann, U. Dirnagl & J. Priller: Bone
marrow-derived cells expressing green fluorescent protein
under the control of the glial fibrillary acidic protein
promoter do not differentiate into astrocytes in vitro and in
vivo. J Neurosci 23(12), 5004-5011 (2003)
106. Vallieres L. & P. E. Sawchenko: Bone marrow-
derived cells that populate the adult mouse brain preserve
their hematopoietic identity. J Neurosci 23(12), 5197-5207
(2003)
107. Castro R. F., K. A. Jackson, M. A. Goodell, C. S.
Robertson, H. Liu & H. D. Shine: Failure of bone marrow
cells to transdifferentiate into neural cells in vivo. Science
297(5585), 1299 (2002)
108. Chen J., Z. G. Zhang, Y. Li, L. Wang, Y. X. Xu, S. C.
Gautam, M. Lu, Z. Zhu & M. Chopp: Intravenous
administration of human bone marrow stromal cells
induces angiogenesis in the ischemic boundary zone after
stroke in rats. Circ Res 92(6), 692-699 (2003)
109. Li Y., J. Chen, X. G. Chen, L. Wang, S. C. Gautam,
Y. X. Xu, M. Katakowski, L. J. Zhang, M. Lu, N.
Janakiraman & M. Chopp: Human marrow stromal cell
therapy for stroke in rat: neurotrophins and functional
recovery. Neurology 59(4), 514-523 (2002)
110. Borlongan C. V., J. G. Lind, O. Dillon-Carter, G. Yu,
M. Hadman, C. Cheng, J. Carroll & D. C. Hess: Bone
marrow grafts restore cerebral blood flow and blood brain
barrier in stroke rats. Brain Res 1010(1-2), 108-116 (2004)
111. Bang O. Y., J. S. Lee, P. H. Lee & G. Lee: Autologous
mesenchymal stem cell transplantation in stroke patients.
Ann Neurol 57(6), 874-882 (2005)
112. Zou Y. R., A. H. Kottmann, M. Kuroda, I. Taniuchi &
D. R. Littman: Function of the chemokine receptor CXCR4
in haematopoiesis and in cerebellar development. Nature
393(6685), 595-599 (1998)
113. Ma Q., D. Jones, P. R. Borghesani, R. A. Segal, T.
Nagasawa, T. Kishimoto, R. T. Bronson & T. A. Springer:
Impaired B-lymphopoiesis, myelopoiesis, and derailed
cerebellar neuron migration in CXCR4- and SDF-1-
deficient mice. Proc Natl Acad Sci USA 95(16), 9448-9453
(1998)
114. Stumm R. K., J. Rummel, V. Junker, C. Culmsee, M.
Pfeiffer, J. Krieglstein, V. Hollt & S. Schulz. A dual role
for the SDF-1/CXCR4 chemokine receptor system in adult
brain: isoform-selective regulation of SDF-1 expression
modulates CXCR4-dependent neuronal plasticity and
cerebral leukocyte recruitment after focal ischemia. J
Neurosci 22(14), 5865-5878 (2002)
115. Imitola J., K. Raddassi, K. I. Park, F. J. Mueller, M.
Nieto, Y. D. Teng, D. Frenkel, J. Li, R. L. Sidman, C. A.
Walsh, E. Y. Snyder & S. J. Khoury: Directed migration of
neural stem cells to sites of CNS injury by the stromal cell-
derived factor 1alpha/CXC chemokine receptor 4 pathway.
Proc Natl Acad Sci USA 101(52), 18117-18122 (2004)
116. Flynn G., S. Maru, J. Loughlin, I. A. Romero & D.
Male: Regulation of chemokine receptor expression in
human microglia and astrocytes. J Neuroimmunol 136(1-2),
84-93 (2003)
117. Croitoru-Lamoury J., G. J. Guillemin, F. D. Boussin,
B. Mognetti, L. I. Gigout, A. Cheret, B. Vaslin, R. Le
Grand, B. J. Brew & D. Dormont: Expression of
chemokines and their receptors in human and simian
astrocytes: evidence for a central role of TNF alpha and IFN
gamma in CXCR4 and CCR5 modulation. Glia 41(4), 354-370
(2003)
118. Oh J. W., K. Drabik, O. Kutsch, C. Choi, A. Tousson &
E. N. Benveniste: CXC chemokine receptor 4 expression and
function in human astroglioma cells. J Immunol 166(4), 2695-
2704 (2001)
119. Kempermann G., H. G. Kuhn & F. H. Gage: Genetic
influence on neurogenesis in the dentate gyrus of adult mice.
Proc Natl Acad Sci USA 94(19), 10409-10414 (1997)
120. Lois C. & A. Alvarez-Buylla: Long-distance neuronal
migration in the adult mammalian brain. Science 264(5162),
1145-1148 (1994)
121. Luskin M. B.: Restricted proliferation and migration of
postnatally generated neurons derived from the forebrain
subventricular zone. Neuron 11(1), 173-189 (1993)
122. Nait-Oumesmar B., L. Decker, F. Lachapelle, V.
Avellana-Adalid, C. Bachelin & A. B. Van Evercooren:
Progenitor cells of the adult mouse subventricular zone
proliferate, migrate and differentiate into oligodendrocytes
after demyelination. Eur J Neurosci 11(12), 4357-4366
(1999)
Neuroprotection by G-CSF
724
123. Kelly S., T. M. Bliss, A. K. Shah, G. H. Sun, M. Ma,
W. C. Foo, J. Masel, M. A. Yenari, I. L. Weissman, N.
Uchida, T. Palmer & G. K. Steinberg: Transplanted human
fetal neural stem cells survive, migrate, and differentiate in
ischemic rat cerebral cortex. PNAS 101(32), 11839-11844
(2004)
124. Jeong S. W., K. Chu, K. H. Jung, S. U. Kim, M. Kim
& J. K. Roh: Human neural stem cell transplantation
promotes functional recovery in rats with experimental
intracerebral hemorrhage. Stroke 34(9), 2258-2263 (2003)
125. Chu K., M. Kim, S.W. Jeong, S.U. Kim & B.W.
Yoon: Human neural stem cells can migrate, differentiate,
and integrate after intravenous transplantation in adult rats
with transient forebrain ischemia Neurosci. Lett 343(2),
129-133 (2003)
126. Takagi Y., K. Nozaki, J. Takahashi, J. Yodoi, M.
Ishikawa & N. Hashimoto: Proliferation of neuronal
precursor cells in the dentate gyrus is accelerated after
transient forebrain ischemia in mice. Brain Res 831(1-2),
283-287 (1999)
127. Zhang R. L., Z. G. Zhang, L. Zhang & M. Chopp:
Proliferation and differentiation of progenitor cells in the
cortex and the subventricular zone in the adult rat after
focal cerebral ischemia. Neuroscience 105(1), 33-41 (2001)
128. Chen X. H., A. Iwata, M. Nonaka, K. D. Browne & D.
H. Smith: Neurogenesis and glial proliferation persist for at
least one year in the subventricular zone following brain
trauma in rats. J Neurotrauma 20(7), 623-631 (2003)
129. Raber J., Y. Fan, Y. Matsumori, Z. Liu, P. R.
Weinstein, J. R. Fike & J. Liu: Irradiation attenuates
neurogenesis and exacerbates ischemia-induced deficits.
Ann Neurol 55(3), 381-389 (2004)
130. Jin K., Y. Sun, L. Xie, J. Childs, X. O. Mao & D. A.
Greenberg: Post-ischemic administration of heparin-
binding epidermal growth factor-like growth factor (HB-
EGF) reduces infarct size and modifies neurogenesis after
focal cerebral ischemia in the rat. J Cereb Blood Flow
Metab 24(4), 399-408 (2004)
131. Wang L., Z. Zhang, Y. Wang, R. Zhang & M. Chopp:
Treatment of stroke with erythropoietin enhances
neurogenesis and angiogenesis and improves neurological
function in rats. Stroke 35(7), 1732-1737 (2004)
132. Zhang R., Y. Wang, L. Zhang, Z. Zhang, W. Tsang,
M. Lu, L. Zhang & M. Chopp: Sildenafil (Viagra) induces
neurogenesis and promotes functional recovery after stroke
in rats. Stroke 33(11), 2675-2680 (2002)
133. Wang L., Z. Gang Zhang, R. Lan Zhang & M. Chopp:
Activation of the PI3-K/Akt pathway mediates cGMP
enhanced-neurogenesis in the adult progenitor cells derived
from the subventricular zone. J Cereb Blood Flow Metab
25(9), 1150-1158 (2005)
134. Katakowski M., Z. G. Zhang, J. Chen, R. Zhang, Y.
Wang, H. Jiang, L. Zhang, A. Robin, Y. Li & M. Chopp:
Phosphoinositide 3-kinase promotes adult subventricular
neuroblast migration after stroke. J Neurosci Res 74(4),
494-501 (2003)
135. Peng H., Y. Huang, J. Rose, D. Erichsen, S. Herek, N.
Fujii, H. Tamamura & J. Zheng: Stromal cell-derived factor
1-mediated CXCR4 signaling in rat and human cortical
neural progenitor cells. J Neurosci Res 76(1), 35-50 (2004)
136. Robin A. M., Z. G. Zhang, L. Wang, R. L. Zhang, M.
Katakowski, L. Zhang, Y. Wang, C. Zhang & M. Chopp:
Stromal cell-derived factor 1alpha mediates neural
progenitor cell motility after focal cerebral ischemia. J
Cereb Blood Flow Metab doi:10.1038/sj.jcbfm.9600172
Key Words: Angiogenesis, Apoptosis, Cytokine,
Granulocyte-colony stimulating factor, Growth factor,
Inflammation, Neurogenesis, Neuroprotection, Plasticity,
Stem cell, Stroke, Review
Send correspondence to: John H. Zhang M.D., Ph.D.,
Division of Neurosurgery, Loma Linda University Medical
Center, 11234 Anderson Street, Room 2562B, Loma Linda,
California 92354, Tel: 909-558-4723, Fax: 909-558-0119,
E-mail: johnzhang3910@yahoo.com
http://www.bioscience.org/current/vol12.htm
... G-CSF applies its neuroprotective features by inhibiting inflammation and apoptosis and stimulating neurogenesis. It also brings together bone marrow stem cells in the damaged brain (Solaroglu et al., 2007, Song et al., 2016. Also, granulocyte colonystimulating factor (G-CSF) is a glycoprotein expressed in neurons that is upregulated by ischemia and prevents programmed cell death in neurons (Schneider et al., 2005). ...
... Also, granulocyte colonystimulating factor (G-CSF) is a glycoprotein expressed in neurons that is upregulated by ischemia and prevents programmed cell death in neurons (Schneider et al., 2005). G-CSF activates Stat family proteins or the PI3-K/Akt pathway and inhibits apoptosis and stimulates cell differentiation in the nervous system (Solaroglu et al., 2007). G-CSF suppresses matrix metallopeptidase 9 (MMP-9), which plays a key role in mediating inflammation, BBB breakdown with subsequent edema formation, and tissue damage in acute stroke (Minnerup et al., 2009). ...
... Following the induction of cerebral ischemia by MCAO, rats were randomly divided to receive G-CSF or vehicle. The treatment group received G-CSF 50 µg/kg subcutaneously (Solaroglu et al., 2007, Minnerup et al., 2008. 6, 24, and 48 h after brain ischemia induction, and the control group received vehicle only. ...
Article
Full-text available
Background Stroke remains the leading cause of death and disability in the world. A new potential treatment for stroke is the granulocyte colony-stimulating factor (G-CSF), which exerts neuroprotective effects through multiple mechanisms. Memory impairment is the most common cognitive problem after a stroke. The suggested treatment for memory impairments is cognitive rehabilitation, which is often ineffective. The hippocampus plays an important role in memory formation. This project aimed to study the effect of G-CSF on memory and dendritic morphology of hippocampal CA1 pyramidal neurons after middle cerebral artery occlusion (MCAO)in rats. Methods Male Sprague-Dawley rats were divided into three groups: the sham, control (MCAO + Vehicle), and treatment (MCAO + G-CSF) groups. G-CSF (50 µg/kg S.C) was administered at 6, 24, and 48 h after brain ischemia induction. The passive avoidance task to evaluate learning and memory was performed on days 6 and 7 post-ischemia. Seven days after MCAO, the brain was removed and the hippocampal slices were stained with Golgi. After that, the neurons were analyzed for dendritic morphology and maturity. Outcomes The data showed that stroke was associated with a significant impairment in the acquisition and retention of passive avoidance tasks, while the G-CSF improved learning and memory loss. The dendritic length, arborization, spine density, and mature spines of the hippocampus CA1 neurons were significantly reduced in the control group, and treatment with G-CSF significantly increased these parameters. Conclusion G-CSF, even with three doses, improved learning and memory deficits, and dendritic morphological changes in the CA1 hippocampal neurons resulted from brain ischemia.
... CSF3 is an essential growth factor regulating the maturation of granulocytes (27). Furthermore, CSF3 is known to stimulate neurogenesis (28) and has neuroprotective functions (29). The decreased CSF3 levels may partially contribute to APOE4's risk to AD. ...
Article
The immune system substantially influences age-related cognitive decline and Alzheimer’s disease (AD) progression, affected by genetic and environmental factors. In a Mayo Clinic Study of Aging cohort, we examined how risk factors like APOE genotype, age, and sex affect inflammatory molecules and AD biomarkers in cerebrospinal fluid (CSF). Among cognitively unimpaired individuals over 65 ( N = 298), we measured 365 CSF inflammatory molecules, finding age, sex, and diabetes status predominantly influencing their levels. We observed age-related correlations with AD biomarkers such as total tau, phosphorylated tau-181, neurofilament light chain (NfL), and YKL40. APOE4 was associated with lower Aβ42 and higher SNAP25 in CSF. We explored baseline variables predicting cognitive decline risk, finding age, CSF Aβ42, NfL, and REG4 to be independently correlated. Subjects with older age, lower Aβ42, higher NfL, and higher REG4 at baseline had increased cognitive impairment risk during follow-up. This suggests that assessing CSF inflammatory molecules and AD biomarkers could predict cognitive impairment risk in the elderly.
Article
Full-text available
Biosimilars can provide choices for patients and may provide cost savings; however, their uptake has been slow in the USA, in part due to limited knowledge. To provide additional confidence in US pegfilgrastim biosimilars, this narrative review compared the safety profiles of biosimilar pegfilgrastims, currently approved or filed for approval in the USA, with the EU- and US-approved reference pegfilgrastims. Headache and bone pain were common to biosimilars and reference products and occurred at a similar incidence. Clinical trial data on the safety profiles of biosimilar pegfilgrastims and reference products have demonstrated similarity and comparability, with no unexpected safety outcomes. Overall, the safety profiles of biosimilar pegfilgrastims and reference pegfilgrastims demonstrated a high degree of similarity and comparability.
Article
Objective: The incidence of childhood overweight and obesity has been increasing in recent years. Immune dysregulation has been demonstrated as a condition related to childhood obesity. Whether the neonatal immune status is related to infant overweight and obesity at 1 year of age is unclear. Methods: To explore the relationship between neonatal cytokines and infant overweight and obesity, we conducted a prospective study in Suzhou Municipal Hospital Affiliated to Nanjing Medical University from 2015 to 2016. 514 neonates were recruited and their dried blood spots were collected after birth. Infants were grouped into normal size groups and overweight and obesity groups based on BMI at 1 year of age. 27 neonatal cytokines levels were compared between the two groups. Results: 370 infants were included in final analysis. Granulocyte colony stimulating factor (GCSF), interleukin-17A (IL17A) and platelet derived growth factor-BB (PDGF-BB) levels were independently associated with childhood overweight and obesity (OR =1.27, 95%CI 1.03, 1.57; OR =1.29, 95%CI: 1.06, 1.60; OR =0.69, 95%CI: 0.49, 0.96). Additionally, neonatal GCSF and IL17A levels were positively associated with increased BMI (β = 0.11, 95%CI: 0.02, 0.19; β = 0.07, 95%CI 0.01, 013) and BMI z-scores (β = 0.10, 95%CI: 0.02, 0.18; β = 0.06, 95%CI 0.01, 0.13). Neonatal PDGF-BB levels were negatively associated with BMI (β = -0.12, 95%CI: -0.23, -0.01) and BMI z-scores (β = -0.12, 95%CI: -0.23, -0.01). The inverse probability weighting (IPW) was performed to account for potential selection bias of this study, and the results were consistent with the above mentioned findings. Conclusions: Neonatal GCSF, IL17A and PDGF-BB levels were correlated with infant overweight and obesity at 1 year of age, suggesting that early life immune status play a significant role of late obesity.
Article
Retinopathy of prematurity (ROP) is a vasoproliferative disorder of the retina and a leading cause of visual impairment and childhood blindness worldwide. The disease is characterized by an early stage of retinal microvascular degeneration, followed by neovascularization that can lead to subsequent retinal detachment and permanent visual loss. Several factors play a key role during the different pathological stages of the disease. Oxidative and nitrosative stress and inflammatory processes are important contributors to the early stage of ROP. Nitric oxide synthase and arginase play important roles in ischemia/ reperfusion-induced neurovascular degeneration. Destructive neovascularization is driven by mediators of the hypoxia-inducible factor pathway, such as vascular endothelial growth factor and metabolic factors (succinate). The extracellular matrix is involved in hypoxia-induced retinal neovascularization. Vasorepulsive molecules (semaphorin 3A) intervene preventing the revascularization of the avascular zone. This review focuses on current concepts about signaling pathways and their mediators, involved in the pathogenesis of ROP, highlighting new potentially preventive and therapeutic modalities. A better understanding of the intricate molecular mechanisms underlying the pathogenesis of ROP should allow the development of more effective and targeted therapeutic agents to reduce aberrant vasoproliferation and facilitate physiological retinal vascular development.
Chapter
A spinal cord injury (SCI) is a very debilitating condition causing loss of sensory and motor function as well as multiple organ failures. Current therapeutic options like surgery and pharmacotherapy show positive results but are incapable of providing a complete cure for chronic SCI symptoms. Tissue engineering, including neuroprotective or growth factors, stem cells, and biomaterial scaffolds, grabs attention because of their potential for regeneration and ability to bridge the gap in the injured spinal cord (SC). Preclinical studies with tissue engineering showed functional recovery and neurorestorative effects. Few clinical trials show the safety and efficacy of the tissue engineering approach. However, more studies should be carried out for potential treatment modalities. In this review, we summarize the pathophysiology of SCI and its current treatment modalities, including surgical, pharmacological, and tissue engineering approaches following SCI in preclinical and clinical phases.KeywordsNeuroprotectionNeuroregenerationScaffoldsSpinal cord injuryStem cellsTissue engineering
Article
Over the past two decades in situ tissue engineering has emerged as a new approach where biomaterials are used to harness the body's own stem/progenitor cells to regenerate diseased or injured tissue. Immunomodulatory biomaterials are designed to promote a regenerative environment, recruit resident stem cells to diseased or injured tissue sites, and direct them towards tissue regeneration. This review explores advances gathered from in vitro and in vivo studies on in situ tissue regenerative therapies. Here we also examine the different ways this approach has been incorporated into biomaterial sciences in order to create customized biomaterial products for therapeutic applications in a broad spectrum of tissues and diseases. Statement of Significance Biomaterials can be designed to recruit stem cells and coordinate their behavior and function towards the restoration or replacement of damaged or diseased tissues in a process known as in situ tissue regeneration. Advanced biomaterial constructs with precise structure, composition, mechanical, and physical properties can be transplanted to tissue site and exploit local stem cells and their micro-environment to promote tissue regeneration. In the absence of cells, we explore the critical immunomodulatory, chemical and physical properties to consider in material design and choice. The application of biomaterials for in situ tissue regeneration has the potential to address a broad range of injuries and diseases.
Chapter
In this chapter the initial discussion is on the prospects for the cytokine tumor necrosis factor–related apoptosis-inducing ligand (TRAIL) and ovarian cancer. Ensuing topics are TRAIL and its mechanism, the tumor necrosis superfamily, platelet-derived growth factor, epidermal growth factor, transforming growth factor, fibroblast growth factor, neurotrophins and nerve growth factor, colony-stimulating factor, erythropoietin, interferon, insulin-like growth factors, and overview. The chapter ends with a summary, a list of references, multiple-choice review questions, and a case-based problem.
Article
Full-text available
We monitor cellular responses to a penetrating wound in the cerebral cortex of adult rat during the first weeks after injury. Two classes of activated mononuclear phagocytes containing acetylated low-density lipoprotein (ac-LDL) receptors appear within hours at the wound site. One type of cell surrounding the lesion edge had thin, delicate processes and is identical in appearance to ramified microglia found in developing brain. Within the lesion, round cells are recognized as blood-borne macrophages when labeled by intravenous injection of carbon particles. Thus, both process-bearing reactive microglia and invading macrophages respond to brain trauma. The greatest number of ac-LDL(+) or nonspecific esterase(+) mononuclear phagocytes appears 2 days after injury within the wound site and are associated with a peak production of the cytokine interleukin-1 (IL-1). Because intracerebral infusion of IL-1 is known to stimulate astrogliosis and neovascularization (Giulian et al., 1988), we examine the time course of injury-induced reactive astrogliosis and angiogenesis. A 5-fold increase in the number of reactive astroglia is found at 3 d and a marked neovascularization at 5 d after injury. During the first week, mononuclear phagocytes engulf particles and clear them from the wound site either by migrating to the brain surface or by entering newly formed brain vasculature. To investigate further the role of reactive brain mononuclear phagocytes in CNS injury, we use drugs to inhibit trauma-induced inflammation. When applied in vivo, chloroquine or colchicine reduce the number of mononuclear phagocytes in damaged brain, help to block reactive astrogliosis and neovascularization, and slow the rate of debris clearance from sites of traumatic injury. In contrast, the glucocorticoid dexamethasone neither reduces the number of brain inflammatory cells nor hampers such responses as phagocytosis, astrogliosis, neovascularization, or debris clearance in vivo. Our observations show that mononuclear phagocytes play a major role in wound healing after CNS trauma with some events controlled by secretion of cytokines. Moreover, certain classes of immunosuppressive drugs may be useful in the treatment of acute brain injury.
Article
Full-text available
In a double-blind, placebo-controlled, randomized study, 10 healthy men received either a single dose of 480 microg granulocyte colony-stimulating factor (G-CSF) or saline. Blood taken from the volunteers was stimulated with 10 microg/mL endotoxin and released cytokines were measured by enzyme-linked immunosorbent assay. Expression of G-CSF receptors on leukocytes was examined by flow cytometry and reverse transcriptase-polymerase chain reaction. Functional activity of these receptors was tested by challenging isolated leukocyte populations to release cytokines with endotoxin in the presence of G-CSF. The G-CSF treatment attenuated the release of the proinflammatory cytokines tumor necrosis factor (TNF)-alpha, interleukin (IL)-12, IL-1beta, and interferon (IFN)-gamma in ex vivo lipopolysaccharide (LPS)-stimulated whole blood. In blood from untreated volunteers the presence of G-CSF in vitro also attenuated the LPS-stimulated release of these cytokines. G-CSF in vitro also attenuated TNF-alpha release from elutriation-purified monocytes. In the presence of 10 ng/mL recombinant TNF-alpha, the attenuation of LPS-inducible IFN-gamma release by G-CSF was blunted in whole blood. However, G-CSF had no such effect on IFN-gamma release from isolated lymphocytes stimulated with anti-CD3 or a combination of TNF-alpha and IL-12. G-CSF receptor expression was detected in human neutrophils and monocytes but not in lymphocytes by means of RT-PCR as well as flow cytometry. These results indicate that G-CSF receptors expressed on monocytes are functional in modulating monokine release. We conclude that the attenuation of IFN-gamma release from lymphocytes is not a direct effect of G-CSF on these cells but is rather due to the inhibition of monocytic IL-12 and TNF-alpha release by G-CSF. (Blood. 2000;95:270-276)
Article
Full-text available
Pretreatment with recombinant human granulocyte CSF (G-CSF) protected mice in two different models of septic shock. Intravenous injection of 250 micrograms/kg G-CSF to mice prevented lethality induced by 5 mg/kg LPS. Injection of 50 micrograms/kg G-CSF protected galactosamine-sensitized mice against LPS-induced hepatitis. In either case, this protection was accompanied by a suppression of LPS-induced serum TNF activity. In contrast, when galactosamine-sensitized mice were pretreated with 50 micrograms/kg murine recombinant granulocyte/macrophage CSF instead of G-CSF and subsequently challenged with LPS, serum TNF activity was significantly enhanced and mortality was increased. The suppressive effect of G-CSF on LPS-induced TNF production was also demonstrated in rats. In vivo, no TNF was detectable in the blood of LPS-treated rats, which had been pretreated with G-CSF. Ex vivo, alveolar macrophages, bone marrow macrophages, Kupffer cells, or peritoneal macrophages prepared from G-CSF-treated rats produced significantly less TNF upon stimulation with LPS than corresponding populations from control rats. However, when these macrophage populations were incubated with G-CSF in vitro, LPS-induced TNF production was unaffected. These data suggest that the G-CSF-mediated suppression of TNF production is not a direct effect of G-CSF on macrophages. To examine whether, independent of the protection against LPS, G-CSF treatment still activated neutrophils, it was demonstrated that granulocytes from G-CSF-treated rats were primed for PMA-induced oxidative burst and for ionophore/arachidonic acid-stimulated lipoxygenase product formation. The experiments of this study support the notion that G-CSF is a negative feedback signal for macrophage-derived TNF-alpha production during Gram-negative sepsis.
Article
In a double-blind, placebo-controlled, randomized study, 10 healthy men received either a single dose of 480 mu g granulocyte colony-stimulating factor (G-CSF) or saline. Blood taken from the volunteers was stimulated with 10 mu g/mL endotoxin and released cytokines were measured by enzyme-linked immunosorbent assay. Expression of G-CSF receptors on leukocytes was examined by flow cytometry and reverse transcriptase-polymerase chain reaction. Functional activity of these receptors was tested by challenging isolated leukocyte populations to release cytokines with endotoxin in the presence of G-CSF. The G-CSF treatment attenuated the release of the proinflammatory cytokines tumor necrosis factor (TNF)-alpha, interleukin (IL)-12, IL-1 beta, and interferon (IFN)-gamma in ex vivo lipopolysaccharide (LPS)-stimulated whole blood, In blood from untreated volunteers the presence of G-CSF in vitro also attenuated the LPS-stimulated release of these cytokines. G-CSF in vitro also attenuated TNF-alpha release from elutriation-purified monocytes, In the presence of 10 ng/mL recombinant TNF-alpha, the attenuation of LPS-inducible lFN-gamma release by G-CSF was blunted in whole blood. However, G-CSF had no such effect on IFN-gamma release from isolated lymphocytes stimulated with anti-CDS or a combination of TNF-alpha and IL-12, G-CSF receptor expression was detected in human neutrophils and monocytes but not in lymphocytes by means of RT-PCR as well as flow cytometry, These results indicate that G-CSF receptors expressed on monocytes are functional in modulating monokine release. We conclude that the attenuation of IFN-gamma release from lymphocytes is not a direct effect of G-CSF on these cells but is rather due to the inhibition of monocytic IL-12 and TNF-alpha release by G-CSF. (C) 2000 by The American Society of Hematology.
Article
There has been an increasing interest during the last ten years or so in the contributions of free radical reactions to the overall metabolic perturbations that result in tissue damage and disease. For a historical overview of free radical-mediated disturbances see Slater (1971); more recent references and comments can be obtained from Slater (1984).
Article
Objective: The objective of this study was to clarify the effects of recombinant human granulocyte colony-stimulating factor (rhG-CSF) administration on infections in patients with severe head injuries after combined therapy with high-dose barbiturates and mild hypothermia. Patients and Methods: Since 1996, we have administered rhG-CSF to eight patients with severe head injuries for 5 days (group G), Their treatment results were compared with those of 22 patients cared for earlier without rhG-CSF treatment (group N), All patients in both groups met the criteria of total leukocyte count (TLC) less than 5,000/mm(3), C-reactive protein (CRP) over 10 mg/dL, and the presence of an infectious complication. Changes in the TLC, CRP, respiratory index, intracranial pressure, and infectious condition were evaluated in both groups. In addition, the nucleated cell count a:nd differentiation from bone marrow aspiration, neutrophil functions, serum concentrations of interleukin-6, and plasma concentration of leukocyte elastase were evaluated in group G, Results: In group G, TLC, nucleated cell count, and neutrophil functions significantly increased, whereas CRP, respiratory index, and interleukin-6 decreased reciprocally. There was no deterioration of intracranial pressure and leukocyte elastase, Consequently, seven of the eight patients in group G recovered from life-threatening infections, and none of the eight patients died. However, in group N, CRP and respiratory index remained high and TLC did not increase as much as it did in group G, Infections continued after 5 days in 17 of the 22 patients, 7 of whom died from severe infections during hospitalization, Conclusion: Administration of rhG-CSF ameliorated life-threatening infections without causing lung injury or increasing brain swelling in patients with severe head injuries who were treated with combined therapy involving high-dose barbiturates and mild hypothermia.
Article
We investigated the proliferation of neuronal progenitor cells by labeling dividing cells by systemic application of the thymidine analog 5-bromodeoxyuridine (BrdU) during transient forebrain ischemia in mice. At 3 (n=6), 7 (n=6), 10 (n=6), and 17 days (n=6) after reperfusion, BrdU-labeled cells were detected in the dentate gyrus and paraventricle lesion. After ischemia-reperfusion, BrdU-labeled cells in the dentate gyrus significantly increased in number but not in the paraventricle lesion. These observations may help to clarify the mechanism of functional recovery after stroke.
Article
Bone marrow stromal cells (BMSC) normally give rise to bone, cartilage, and mesenchymal cells. Recently, bone marrow cells have been shown to have the capacity to differentiate into myocytes, hepatocytes, and glial cells. We now demonstrate that human and mouse BMSC can be induced to differentiate into neural cells under experimental cell culture conditions. BMSC cultured in the presence of EGF or BDNF expressed the protein and mRNA for nestin, a marker of neural precursors. These cultures also expressed glial fibrillary acidic protein (GFAP) and neuron-specific nuclear protein (NeuN). When labeled human or mouse BMSC were cultured with rat fetal mesencephalic or striatal cells, a small proportion of BMSC-derived cells differentiated into neuron-like cells expressing NeuN and glial cells expressing GFAP.
Article
Astrocyte-enriched populations were established from human embryonic brain analyzed for their ability to synthesize cytokines potentially relevant for mechanisms of inflammation and immunity in the brain. Unstimulated astrocytes did not secrete significant IL-6, IL-8, macrophage CSF (M-CSF), granulocyte-macrophage CSF (GM-CSF), or granulocyte-CSF (G-CSF), as determined by specific ELISA and/or bioassay. With the exception of M-CSF mRNA, transcripts for the above factors were not detected in unstimulated astrocytes. On exposure of human astrocytes to IL-1 beta, high levels of IL-6, IL-8, M-CSF, G-CSF, and GM-CSF mRNAs were detected; moreover, active secretion of all the above cytokines was demonstrated. TNF-alpha was also able to stimulate IL-6, IL-8, M-CSF, GM-CSF, and G-CSF synthesis and secretion, but was generally less potent than IL-1 beta. No IL-3 mRNA or protein was detected in unstimulated or cytokine-treated astrocytes. IL-1 alpha and IL-1 beta mRNAs and proteins were not detected in unstimulated astrocytes, but were present in very small amounts after stimulation with TNF-alpha/IL-1 beta. No IL-6, M-CSF, GM-CSF, G-CSF, or IL-8 were induced by IL-1 beta or TNF-alpha in early primary cultures, which mainly contain undifferentiated neuronal/glial progenitor cells. These studies demonstrate for the first time the production of multiple cytokines by normal human astrocytes stimulated in culture by IL-1 beta and TNF-alpha. The capacity of human astrocytes to synthesize and release cytokines active on hemolymphopoietic cells supports the concept that these cells play an important role in the regulation of inflammatory and immune responses in a variety of brain pathologies.