ArticlePDF AvailableLiterature Review

Abstract

Abstract Neurological disorders, such as stroke, are triggered by a loss of neurons and glial cells. Ischemic stroke remains a substantial problem for industrialized countries. Over the previous few decades our understanding about the pathophysiology of stroke has enhanced, nevertheless, more awareness is required to advance the field of stroke recovery. Existing therapies are incapable to adequately relief the disease outcome and are not appropriate to all patients. Meanwhile, the majority of patients continue to show neurological deficits even subsequent effective thrombolysis, recuperative therapies are immediately required that stimulate brain remodeling and repair once stroke damage has happened. Cell therapy is emergent as a hopeful new modality for increasing neurological recovery in ischemic stroke. Numerous types of stem cells from various sources have been identified and their possibility and efficiency for the treatment of stroke have been investigated. Stem cell therapy in patients with stroke using adult stem cells have been first practiced in clinical trials since 15 years ago. Even though stem cells have revealed a hopeful role in ischemic stroke in investigational studies besides early clinical pilot studies, cellular therapy in human is still at a primary stage. In this review, we summarize the types of stem cells, various delivery routes, and clinical application of stem cell‐based therapy for stroke treatment.
Received: 29 August 2018
|
Accepted: 15 November 2018
DOI: 10.1002/jcb.28207
PROSPECTS
Stem cellbased cell therapy for neuroprotection in stroke:
A review
Reza Rikhtegar
1
|
Mehdi Yousefi
2,3
|
Sanam Dolati
1,4
|
Hosein Delavar Kasmaei
5
|
Saeid Charsouei
1
|
Mohammad Nouri
2
|
Seyed Kazem Shakouri
1,6
1
Aging Research Institute, Tabriz
University of Medical Sciences,
Tabriz, Iran
2
Stem Cell Research Center, Tabriz
University of Medical Sciences,
Tabriz, Iran
3
Department of Immunology, School of
Medicine, Tabriz University of Medical
Sciences, Tabriz, Iran
4
Student's Research Committee, Tabriz
University of Medical Sciences,
Tabriz, Iran
5
Department of Neurology, Shohadae
Tajrish Hospital, Shahid Beheshti
University of Medical Sciences,
Tehran, Iran
6
Physical Medicine and Rehabilitation
Research Centre, Tabriz University of
Medical Sciences, Tabriz, Iran
Correspondence
Mehdi Yousefi, PhD, Assistant Professor
of Immunology, Department of
Immunology, Faculty of Medicine, Tabriz
University of Medical Sciences, Tabriz,
Iran.
Email: Yousefime@tbzmed.ac.ir
Seyed Kazem Shakouri, M.D. Professor,
Department of Physical Medicine &
Rehabilitation, Tabriz University of
Medical Sciences, Tabriz, Iran.
Email: shakourik@tbzmed.ac.ir
Funding information
Aging Research Institute, Tabriz
University of Medical Sciences
Abstract
Neurological disorders, such as stroke, are triggered by a loss of neurons and glial
cells. Ischemic stroke remains a substantial problem for industrialized countries.
Over the previous few decades our understanding about the pathophysiology of
stroke has enhanced, nevertheless, more awareness is required to advance the field of
stroke recovery. Existing therapies are incapable to adequately relief the disease
outcome and are not appropriate to all patients. Meanwhile, the majority of patients
continue to show neurological deficits even subsequent effective thrombolysis,
recuperative therapies are immediately required that stimulate brain remodeling and
repaironcestrokedamagehashappened.Celltherapyisemergentasahopefulnew
modality for increasing neurological recovery in ischemic stroke. Numerous types of
stem cells from various sources have been identified and their possibility and
efficiency for the treatment of stroke have been investigated. Stem cell therapy in
patients with stroke using adult stem cells have been first practiced in clinical trials
since 15 years ago. Even though stem cells have revealed a hopeful role in ischemic
stroke in investigational studies besides early clinical pilot studies, cellular therapy in
human is still at a primary stage. In this review, we summarize the types of stem
cells, various delivery routes, and clinical application of stem cellbased therapy for
stroke treatment.
KEYWORDS
clinical trials., ischemic stroke, stem cell therapy
1
|
INTRODUCTION
Stroke is one of the most common causes of the death,
after cancer and myocardial infarction and happens
mainly in the old population with a higher risk in men.
1
Reports indicate that the morbidity and mortality of
stroke have increased during previous decades.
2
Globally,
stroke is in second or third place of mortality list and the
fifth principal reason of death in the United States.
3
Stroke is triggered by stumbling block of a cerebral artery,
leading to focal ischemia, loss of neurons and glial cells,
causing motor, sensory, or cognitive injuries. Two
J Cell Biochem. 2018;1-14. wileyonlinelibrary.com/journal/jcb © 2018 Wiley Periodicals, Inc.
|
1
foremost types of stroke are known: Hemorrhagic and
ischemic stroke.
4
Hemorrhagic stroke happens as soon as
rupture of blood vessels in the brain, whereas ischemic
stroke occurs following embolism, thrombolysis, or
cryptogenic mechanisms in which blood supply to the
brain is disturbed and is described as the main type of
ischemic stroke (87%).
5
Ischemic stroke happens when
an area of brain tissue is underprivileged of oxygen
supply because of a reduction in local blood flow. The
normal function of the brain ends if oxygen deprivation
exceeds 60 to 90 seconds and brain tissue death happens
within 3 hours of anoxia leading to cerebral infarction.
5,6
The severe injury to the brain tissues following ischemic
stroke contains not only devastation of a heterogeneous
population of brain cell types, but also main damage of
neuronal networks and vascular systems.
7
It has been
known that brain ischemia motivates neurogenesis by
stimulating neuronal migration through the damaged
area via secretion of neurotrophic factors such as brain
derived neurotrophic factor (BDNF), vascular endothelial
growth factor (VEGF), cytokines like monocyte chemoat-
tractant protein (MCP1), and macrophage inflammatory
protein (MIP1).
5
Even though there are developing
treatments that repair perfusion to the ischemic brain,
containing tissue plasminogen activator (tPA), a throm-
bolytic agent, can only be administered during the acute
onset of stroke pathology,
8
along with intraarterial/
endovascular processes to recanalize blood vessels.
These types of treatment reported to decrease stroke
mortality.
9,10
Currently, the only confirmed therapy for
ischemic stroke is thrombolysis, which must be applied
within 4.5 hours after attack.
11
However, due to hemor-
rhagic problem, thrombolysis is still not commonly
practiced.
12
Furthermore, accessible surgical interven-
tions aim to decrease the general risk of clot formation
during stroke.
13
Neurological injury as a consequence of
ischemia is in principal permanent, thus it is essential to
progress innovative therapeutic attitudes to recover
missing neurological functions over the renewal of
neurons.
14
Available treatment possibilities for patients
with ischemic stroke are limited. Therefore, the perfect
cellbased therapies should not only directed towards
replacement of missing cell types, renovation of func-
tional, and suitable neuronal networks, but also the
rebuilding of disturbed vascular systems.
15,16
Human
cells should be capable of substituting dead neurons,
remyelination of axons, and repair of injured neural
circuitries.
17
This review presents the most current
improvements in stem cell therapy applied for ischemic
stroke, aiming to evaluate its safety and efficiency profile
with emphasis on embryonic stem cells (ESCs), me-
senchymal stem cells (MSCs), and neural stem cells
(NSCs).
2
|
PATHOPHYSIOLOGY OF
ISCHEMIC STROKE
After an ischemic stroke, neurons are left without oxygen
and energy whereas, energydependent processes in neuro-
nal cells are greatly affected.
18
Instantly following ischemia,
neurons fail to maintain their normal transmembrane ionic
gradient and homeostasis properties.
19
This phenomena
could induce numerous processes causing the cell death,
such as excitotoxicity, mitochondrial dysfunction, inflam-
mation, and oxidative and nitrative stress, because of a great
intracellular influx of Ca
2+
ions subsequent disruption of
transmembrane protein channels. Moreover, production of
reactive oxygen species (ROS) upregulated by Ca
2+
influx in
the mitochondria implicated in reperfusion damage after
ischemia leading to necrosis.
20
In addition, due to a
disruption in the ionic gradients following ischemia,
availability to nutrients required for neuronal cells is
reduced which leads the overproduction of excitatory
amino acids such as glutamate.
21-23
NmethylDaspartate
(NMDA) glutamate receptor prompts augmented amounts
of intracellular Ca
2+
influx and leads the activation of Ca
2+
dependent enzymes comprising proteases, calpain, and
caspases dependent cellular death pathways containing
caspase12, caspase9, and caspase3 after the release of
cytochrome C, thereby setting off mitochondrial mechan-
isms of apoptosis and necrosis.
24
Dying neural cells could
release signals to activate proinflammatory pathways
leading to post ischemic inflammation that plays a role to
trigger the immune response. Both proand antiinflam-
matory mediators are described to implicate in the
pathogenesis of ischemic stroke.
25
Inflammatory cells are
contributed to ischemic strokerelated inflammation; pro-
cedures that are associated with the act of interleukin17A
(IL17A).
7
In the cerebrospinal fluid (CSF) of patients in the
acute period of ischemic hemispheric stroke, proinflamma-
tory cytokines are raised; while, protective antiinflamma-
tory and trophotropic factors are reduced, which may
stimulate an inflammatory response after ischemic stroke.
25
3
|
STEM CELLBASED
THERAPIES
Stem cells are undifferentiated cells capable of self
renewal and proficient of differentiation into multiple
cell types. Stem cell therapy has gained consideration
increasingly as a treatment of unmanageable disorder
such as ischemic stroke,
26
possibly by secreting various
neural trophic factors, immunomodulation, neuroprotec-
tion, angiogenesis, and maybe also prompting neuronal
replacement.
27
The first aim of implementing stem cells
to treat ischemic stroke was to renew the strokeimpaired
2
|
RIKHTEGAR ET AL.
tissue using cellular replacement. Locally injection of
cells to the ischemic area has shown to be more effective
than the intravenous perfusion.
28
Amazingly, various
investigators established that nearly 80% of transferred
cells die 3 days post transplantation, due to the
antagonistic microenvironment of the lesion site.
28
Transplantation of stem cells, causes the immune
response,
29
and immune rejection is still a main draw-
back mostly in the clinical studies of neurological
disorders in which immune suppression is part of the
clinical procedure protocol.
30
Use of biocompatible and
biodegradable biomaterials including Alginate, Dextran,
And Hyaluronan/methyl cellulose (HAMC), in which are
not generate major immune response, is a hopeful
attitude for facilitation of the effectiveness of trans-
planted stem cells in stroke treatment. Numerous clinical
trials investigating the effect of stem cell therapy for
stroke patients are being conducted. The outcome hope-
fully will provide indication on the therapeutic effect of
cell transplantation, however, the effectiveness of stem
cell therapy in greater numbers of stroke patients is yet to
be confirmed.
31,32
Schematic presentation of stem cell
therapy in stroke is illustrated in Figure 1.
3.1
|
Embryonic stem cells
Embryonic stem cells (ESCs) are pluripotent cells
retrieved from the internal cell bulk of human blasto-
cysts, the inner cell mass (ICM). ESCs have the strong
ability to limitless selfrenewal further discriminate into
cells of all three germ layers, that is, endoderm,
mesoderm, and ectoderm.
33
In particular settings, after
administration of fibroblast growth factor2 (FGF2),
ESCs could be differentiate into neural lineage cell
types.
34
The differentiated neural cells have been
reported to express glutamatergic, GABAergic, or dopa-
minergic markers.
34
Humanderived ESCs have been
broadly investigated during current years for generation
of different types of neurons.
35,36
ESCsderived mesench-
ymal stem cells, vascular progenitor cells, and neural
progenitor cells have been revealed to provide valuable
effects without visible tumorigenesis.
37,38
Neuronal pro-
genitor cells derivative from ESCs are capable of
decreasing infarct volume, rendering neurogenesis, and
improving developmental behavior.
39
Transferred em-
bryonic neural stem cells are able to motivate angiogenic
cytokines, through vascular endothelial proliferation
within 15 days post cerebral ischemia.
40
To date no
FIGURE 1 The schema picture of stem cells therapy in brain stroke. Stem cells separated from various source tissues and proliferated,
differentiated or genetically modified in in vivo environment. In the second stage stem cells administrated in various routes such as
intravenous, intrathecal, and intraperitoneal. Finally, this stem cells induce their therapeutic effects in different manners; 1. Direct
differentiation to neuron, oligodendrocyte, and astrocyte. 2. Stimulation of angiogenesis. 3. Stimulation of synaptic plasticity and new
synaptic formation. 4. Stimulation of endogenous neurogenesis. NSC migration and proliferation. BM; bone marrow. UCB; umbilical cord
blood. AD; adipose tissue. HSC; hematopoietic stem cell. ESC; embryonic stem cell. MSC; mesenchymal stem cell. iPSC; induced pluripotent
stem cell. NSC; nero stem cell. CNS: central nervous system. IV, intravenous; IT, Intrathecal; IP, Intraperitoneal
RIKHTEGAR ET AL.
|
3
clinical trials have been implemented on the use of ESCs
for stroke treatment. Objections such as ethical concerns,
heterogeneity of donor cells, immunological response,
and restricted accessibility of ESCs, restrict their possible
usage for clinical applications. Nevertheless, more basic
and preclinical experimentation should be performed to
understand the possible clinical applications of ESCs.
40,41
3.2
|
Mesenchymal stem cells
Mesenchymal stem cells (MSCs) are multipotent cells,
derived from bone marrow, heart and adipose tissue,
placenta, and skeletal muscle, which are generally
scattered in the entire body and are comfortable to
isolate, with the exceptional capability to differentiate
into mesodermal, endodermal, and ectodermal cell types,
including neurons.
42,43
MSCs are proficient to cross the
bloodbrain barrier (BBB) and specially migrate to the
injured sites, slow down the apoptosis process, raise basic
fibroblast growth factor, and stimulate endogenous
cellular proliferation.
44
The extensive accessibility of
MSCs, creates them a worthy cell candidate for ther-
apeutic approaches concerning stroke.
45
Transplantation
of MSCs into middle cerebral artery occlusion (MCAO) of
stroked rats has been reported to cause the motor
recovery through enhancing of the angiogenesis, and
upregulation of cellular plasticity.
46
MSCs could further
stimulate stroke recovery through mediation of secretion
of neurotrophic factors, such as brain derived neuro-
trophic factor (BDNF) and also lead to secret the
angiogenic mediators.
47
Systemic or peripheral adminis-
tration of MSCs have been reported as a safe and
operative practice for stem cell transplantation.
48
Intra-
venous administration of allogenic MSCs has been
confirmed functional for recovery following stroke.
49
MSCs can be obtained from various peripheral tissues of
an individual. Therefore, autologous MSCs are another
probable source of stem cells that produce a minus severe
immune reaction subsequent transplantation.
50
While
use of autologous MSCs is the method of select to guard
against immune rejection, the elongated time frame
required to attain adequate numbers of MSCs from the
patient's own tissue, makes the usage of offtheshelf
allogeneic MSC therapy more suitable.
51
3.3
|
Neural stem cells
Neural stem cells (NSCs) play a significant role in brain
homeostasis and have been known to imply therapeutic
activities subsequent neurovascular damage.
52
NSCs may
be derived from embryonic, fetal, or adult brain and have
the capability to form all cell types necessary to advance
neurological function.
27
NSCs transplantation is an
efficacious therapy for ischemic stroke over numerous
mechanisms, such as maintenance of the BBB, lessening
of neuroinflammation, increased neurogenesis and angio-
genesis, and eventually practical neurological recovery.
53
Human neural progenitor cells (hNPCs) derived from
embryonic and fetal tissues, have the capability to create
neurons, astrocytes, and oligodendrocytes, as well as
integrating to the host tissue and establish neuronal
features, such as synapse formation, expression of synaptic
proteins, and electrophysiological properties.
54
Brain
ischemia could increase endogenous hNPCs and occa-
sionally induce differentiation into the principal cell types
of the damaged site.
55,56
Furthermore, neuroinflammation
subsequent ischemic stroke is competent to prompt NSCs
enrollment.
57,58
Other possible strategies to rise endogen-
ous NPCs proliferation is administration of hormones such
as erythropoietin, and antiinflammatory drugs such as
indomethacin.
59
Stem cells function as a local or systemic
immunoregulatory machinery and could motivate the
recovery after stroke through diminishing inflammatory
molecules.
60
hNPCs might improve poststroke plasticity
by enhancing of synapse creation, dendritic branch off,
and axonal connections.
61
3.4
|
Other stem cells
3.4.1
|
Induced pluripotent stem cells
Induced pluripotent stem cells (iPSCs), are another
source of stem cells and express four critical factors, that
is Oct3/4, Sox2, cMyc, and Klf4.
62
Human induced
pluripotent stem cells (hiPSCs) encourage possible
restorative abilities after ischemic stroke over their
neuroprotective and neurodegenerative features.
63
Dur-
ing recent years, iPSCs have gained increasing attention
as an interesting cell source for reparation of neuronal
network disturbed by ischemic stroke, however, the
tumorgenicity of grafted iPSCs is still a serious pro-
blem.
64,65
iPSCderived tumors have shown to have
higher expressions of phosphorylated vascular endothe-
lial growth factor receptor2(pVEGFR2) and matrix
metallo proteinase9 (MMP9), which might be involved
in stimulating the teratoma creation.
66
Human fibroblast
derived iPSCs grafted to rats subsequent MCAO stroke
transfer to the injured zone and partly reinstate
sensorimotor function.
67
Promptly, proliferation, and
multipotential differentiation of iPSCs imply that these
type of potent cells could be very hopeful after stroke
complications.
67
Considering the pros and cons of
application of iPSCs as an autologous source for patients
with stroke, there are numerous factors which requisite
to be taken into account. First, the risk for stroke in 75 to
84 years old is 25fold higher than 45 to 54yearsold
people.
68
Second, the vast majority of stroke patients are
4
|
RIKHTEGAR ET AL.
older than 75 years, so generation of iPSCs from aged
sources would be challenging. And finally, the effective
generation and expansion of iPSCs from an aged patient's
skin fibroblast cells based on present technologies is not
possible.
69
Generation of wellcharacterized iPSCs toward
desired a neuronal phenotype, with adequate number
required for transplantation might take at least 7
weeks.
63,69,70
It is possible that the most appropriate time
for transplantation after stroke in humans will be from
several weeks up to 3 months.
70
iPSCs can make available
immune reactionfree and specially tailored stem cell
therapy.
29
The administration of iPSCs derived from the
somatic cells of the transplant recipient, could overcome
the immune reaction.
71
Establishment of iPSC banks has
been considered in numerous countries such as Japan,
the United States, and the United Kingdom. The most
advanced iPSC bank is located in Japan and by 2022 is
expected to have about 60 iPSC lines covering all human
leukocyte antigen haplotypes for the entire population of
Japan.
72
3.4.2
|
Hematopoietic stem cells
Hematopoietic stem cells (HSCs) are another class of
stem cells derived from bone marrow, and are capable to
differentiate into red blood and lymphoid cells.
73
Administration of HSCs has been reported to diminish
the ischemic infarct volume at cerebral cortex of the
MCAO stroke model.
74
Application of HSCs in existence
of stem cell factor (SCF) and granulocytecolony stimu-
lating factor (GCSF) in the hypoxiaischemia model,
reduced atrophy in the ipsilesional cerebral hemi-
sphere.
75
These results imply that HSCs are possibly
valuable stem cells source and valuable candidate for
improving ischemic stroke motivated degeneration.
75
3.4.3
|
Bone marrow stromal cell
Bone marrow stromal cell (BMSCs) express a widerange
of angiogenic/arteriogenic cytokines such as placental
growth factor (PIGF), basic fibroblast growth factor 2
(bFGF/FGF2), vascular endothelial growth factor
(VEGF), insulinlike growth factors (IGFs), and angio-
poietin 1(Ang1),
76,77
that involved in brain plasticity and
retrieval of neurological function following stroke.
78
3.4.4
|
Human umbilical cord blood cells
Human umbilical cord blood cells (HUCBCs) could
mainly differentiate to neurons and a minor group could
differentiate to astrocytes. HUCBCs are consist of
mesenchymal stem cells which differentiate to neural
cells, and also contain a big amount of hematopoietic
colonyforming cells.
79,80
Systemic administration of
CD34+ cells derived from HUCBCs following stroke
stimulated angiogenesis and neurogenesis, and developed
behavioral recovery.
81
Administration of HUCBCs sub-
sequent cerebral ischemia has been known to reduce
neuroinflammation
82
through enhancing the production
of IL10 and reducing of interferonγ(IFNγ), so causing
repression of Tcell proliferation.
83
Primary intravenous
treatment with HUCBCs at 24 hours after MCAO,
improved functional recovery and cell migration, there-
fore, seems to be optimum for clinical treatment of
stroke.
84,85
Although cord blood is introduced as a good
source for cellbased therapies, however, its application
and safety is yet to be confirmed.
86
3.4.5
|
Endothelial progenitor cells
Endothelial progenitor cells (EPCs) are commonly
produced and sustained in bone marrow and could be
transferred into the injury site and contribute into blood
vessel remodeling and repair.
87
Current studies illu-
strated that EPCs transplantation prompted focal angio-
genesis and neurogenesis, improved cerebral blood flow,
diminished neuronal cell death, decreased infarct vo-
lume, and enhanced neurobehavioral retrieval after
ischemia.
87,88
These features of EPCs imply their
therapeutic prospective for treatment of cerebral ische-
mia, and might contribute to blood vessels formation and
release of paracrine trophic factors.
89
In the case of
stroke, different types of cell transplantation, such as
NSCs, MSCs, iPSCs, BMSCs, and HUCBCs have been
described to reduce postischemic inflammation.
90,91
Achieving the mature neuronal phenotype seems to
depend on the source of the stem cells 30% of ESCs, 2% to
20% of MSCs, 34% to 60% of NSCs, 40% to 66% of iPSCs,
and 16.8% of BMSCs could differentiated into neurons.
16
4
|
CELL DELIVERY ROUTES
The effectiveness of stem cell therapy is considerately
linked to the route and location of grafting. Intrapar-
enchymal cell transplantation have shown intense side
effects containing motor deteriorating, syncope, seizures,
and chronic subdural hematoma.
92
In intracerebroven-
tricular delivery, some stroke patients have fever and
meningeal signs following cell transplantation.
93
It was
primarily thought that intracerebral administration was
the best way for exogenous neural stem cells to reach the
brain.
94
Intracerebral administration presented im-
planted cells in the lesion size in comparison with other
delivery routes because several million cells are trans-
planted into the brain and approximately 1/3 of the stem
RIKHTEGAR ET AL.
|
5
cells migrate toward the damaged regions as well as to
the intact hemisphere.
95
Some clinical trials have used
this intracerebral route during the delayed phases for the
reason that it is safer and more suitable for clinical
applications.
94
Intracranial administration of stem cells,
mainly for MSCs, the transport of stem cells into the
circulation system, offers a smaller amount invasive
treatment. Though, any trials procedure should consider
precautions to eliminate the creation of embolisms
following the cell implantation.
96
Intravenously admini-
strated cells go over the systemic and pulmonary vascular
systems, and settle noticeably at the injured site of
brain.
97
After intravenous administration cells may stick
together and cause microemboli, comprising lethal
pulmonary emboli. The intravenous route is more
possible, suitable, and justified cell therapy practice
during the acute or initial subacute time window of
stroke.
98
In fact, the most ongoing clinical trials use
intravenous administration. Intraarterial delivery, by-
passes the peripheral filtering organs, causing to higher
cell engraftment to the brain, and has the superior
effectiveness compared with intravenous infusion.
98
After intraarterial delivery, cells may decrease in
cerebral blood flow related to microstrokes.
99,100
Intra
arterial route of delivery is superior to the intravenous
route for the reason that the cells would be directly
delivered to the brain where they could act to decline
infarct size and rise functional recovery.
101
Intranasal
transport has arisen as a new method to transfer
therapeutic mediators to the brain through the BBB.
This technique is noninvasive and eases cell homing to
the CNS and decreases the possible side effects related to
intravascular administration.
102,103
More progressive de-
livery techniques are also being evaluated, comprising
bioengineered polymers to improve stem cell survival and
effectiveness. Inert polymer matrices, such as hydrogels
and particles, were first applied for stem cell delivery.
104
The various delivery routes to transport the cells to the
stroke region have summarized in Table 1.
5
|
CLINICAL TRIALS OF STEM
CELLS TO STROKE
Literature review revealed that numerous clinical trials
are being conducted on therapeutic effects of stem cells
for stroke. Ten clinical trials of intravascular delivery of
the cells, containing 136 subjects, have been described to
date.
51
The immortalized cell line of teratocarcinoma
derived Ntera2/D1 neuronlike cells (NT2N) were the
primary human cells applied in a Phase I clinical trial for
patients with stroke and were transplanted into the
infarcted area of 12 subjects, six months to six years
afterward a basal ganglia stroke.
105
Functional develop-
ment was seen in this small group of patients without any
important adverse events. In a successive Phase II trial,
NT2N cells were transplanted into the periinfarct or
perihemorrhagic cavity and again promising results
were seen without any unfavorable events.
106
In an
openlabel, singleblinded randomized trial, 50 to 60
million autologous MSCs suspended in 250 mL of saline
were infused intravenously between 3 months and 2
years after stroke. This study presented important
TABLE 1 Routes of cell administration for treatment of stroke
Delivery route Features
Intraparenchymal Early neural transplantation studies
Safety and feasibility, but not
efficacy Cause to:
Motor worsening
Syncope
Chronic subdural hematoma
Intracerebroventricular For NSC transplantation Cause to:
Fever
Meningeal signs
Intracerebral Invasiveness
Poor cell availability
Immune rejection
Uncertain fate in the brain
Development in neurological
consequences
Intracranial Mainly for MSCs transplantation
Less invasive
Intravenous Modest invasiveness
Promote the extensive secretion of
neuroprotective, proangiogenic,
and immunomodulatory factors
Reduce infarct size
Induce functional (motor and/or
cognitive) recovery
Majority of cells become trapped in
the lung, liver, and spleen
Cells may stick together and cause
microemboli
Intraarterial Higher cell engraftment to the
brainmodest invasiveness
Promote the extensive secretion of
neuroprotective, proangiogenic,
and immunomodulatory factors
Decrease in cerebral blood flow
Intranasal Noninvasive method
Poor cell engraftment
Eases cell homing to the CNS
Intrathecal For UCMSCs transplantation
Improve motor function
Decrease ischemic damage of
stroke
Abbreviations: MSCs, mesenchymal stem cells; NSC, neural stem cells;
UCMSCs, umbilical cord mesenchymal stem cells.
6
|
RIKHTEGAR ET AL.
improvement in functional consequence according to the
Modified Rankin Scale (mRS) in the treatment group.
107
In this study, the lesion volume was decreased by 20%
and extraordinarily, no tumorigenesis and venous embo-
lism were revealed. The outcome clearly illustrated the
hopeful possible use of MSCs in clinical settings.
107
In a
SanBioPhase I study, altered donor human BMderived
MSCs (Notchtransfected mesenchymal stromal cells
(SB623 cells) to enhance cell viability) was used. Eighteen
male or female patients were transplanted with 2.5, 5, or
10 million cells between 6 and 60 months (average 22
months) following ischemic stroke.
108
Neurological
function and motor scales above the first 2 to 3 months
post transplantation displayed modest progresses and
were sustained up to 12 months in 16 of the 18 subjects.
This study described the development of hyperintensities
around the needle tracts on T2weighted fluid attenuated
inversionrecovery (FLAIR) scans in most of the patients.
This phenomena was observed one week after transplan-
tation in the SanBio study. However, higher range of T2
hyperintensity was described to associated with develop-
ment in motor damage at 12 months according to the
FuglMeyer motor scale.
108,109
After the hopeful results of
SanBio's earlyphase trial implementing SB623 cells in
chronic stroke, a phase IIb study of a doubleblind, sham
surgery randomized controlled trial, ACTISSIMA (Allo-
geneic Cell Therapy for Ischemic Stroke to Improve
Motor Abilities) has been registered to examine the
effects of stereotactic intracranial implantation of SB623
cells in patients with fixed motor deficits caused by
ischemic stroke.
110
In this study 156 patients from 65
sites in the United States will be enrolled and randomize
patients will be divided into two groups. First group of
patients will receive 2.5 million or 5.0 million SB623 cells,
whereas, the second group subjects will get administrated
by sham placebo injections. The proposed time frame for
this study has been set for 1 year with the primary end
point at 6 months. Patients will be followed for an
additional 6 months. The proportion of patients whose
FuglMeyer motor function total score advances by 10
points at month 6 compared with the control group will
illustrate the effectiveness of the treatment of ACTISSI-
MA study.
110
In the ISISHERMES trial, autologous
MSCs injected intravenously in 31 subacute stroke
patients. This study has also been finalized, nevertheless,
the date is not yet accessible.
111
In a randomized
controlled doubleblinded phase /clinical trial study,
intravenous administration of autologous BMderived
MSC in patients with chronic stroke was performed and
the recovery from hemorrhage were confirmed after the
one year of MSC treatment.
112
Now, accessible data on
clinical trials of MSC therapy indicate that this treatment
improves neurorestoration by rise of neural plasticity and
reduction of lesion volume.
113,114
In a large multicenter
academically funded European trial, Regenerative Stem
Cell Therapy for Stroke in Europe (RESSTORE), adipose
derived donor MSCs transported intravenously 14 days
after ischemic stroke for a group of 400 patients.
109
The
data from this study, however, has not yet published. In
another clinical trial on stem cell therapy for acute
stroke, bone marrow mononuclear cells (BMMNCs)
improved the clinical consequences through a decrease
in the National Institute of Health Stroke Scale (NIHSS)
score six months after transplantation.
115
Clinical trials
using BMMNCs have revealed its safety and practicability
in the acute and chronic phases of recovery.
116
A phase 1/
2A study, which implemented human altered bone
marrowderived stromal cells, presented safety and
feasibility of direct intracerebral implantation six months
to five years after stroke, and also progress in neurolo-
gical consequences.
117
In another clinical trial, the
intrathecally transferred autologous BMMNCs in patients
with chronic stroke, developed the prognosis of practical
recuperation.
118
Intravenous autologous BMMNCs trans-
plantation enhanced the cerebral blood flow and led to
neurological improvement.
119
A Phase 2 randomized,
controlled trials with blinded endpoint valuation took
place at five centers in India. In this study, autologous
BMMNCs transported intravenously at average of 18 days
afterward stroke onset.
120
Sixty patients were assigned to
control and 60 patients to celltherapy injection groups.
Approximately 281 million cells were infused, of which
about 1% was CD34
+
cells. No alterations in functional
consequences were observed through 6 months after
followup.
120
In a recent clinical trial, the safety and
effectiveness of 3 × 10
6
or more autologous BMMNC
transfer in patients with stroke were administrated and
improved consequence in patients with stroke were
accounted.
121
The MASTERS trial was a randomized
double blind dose escalation trial evaluating allogeneic,
adultderived stem cells (MultiStem, Athersys) in the
treatment of early cortical strokes.
122
In randomized
Athersys MultiStem study, intravenous infusion of
allogeneic multipotent adult progenitor cells (MAPCs),
(multipotent bone marrow derivative cells defunct of
CD45
+
/glycophorinA
+
cells) were administrated,
48 hours after onset of ischemic stroke and patients were
then followed for 6 months.
123,124
In this trial, 126
participants (65 and 61 for MAPCs therapy and placebos
respectively) were involved. The MAPCs group showed a
tendency to improved functional consequences within
36 hours post treatment.
125
MultiStem reduces immune
activation and inflammatory responses whereas increases
neurogenesis and differentiation. Decrease in spleen size
that happens subsequent stroke is prohibited
through MultiStem administration along with immune
RIKHTEGAR ET AL.
|
7
TABLE 2 Major clinical trials investigated stem cells therapies for stroke treatment
Cell type Cell source Patient characteristics
Cell volume and
administration time Outcome Adverse effects Reference
1 LBS neurons
(Layton Bioscience
Phase I)
Immortalized cell lines
Ntera2/D1 NeuronLike
Cells (NT2N)
12 Ischemic stroke patients
(6 m6 y after stroke)*
2 Million cells, 6 million
cells
Feasible Kondziolka
et al
105
Age: 4475 y Intracerebral Improve function
2 LBS neurons
(Layton Bioscience
Phase II)
Immortalized cell lines
Ntera2/D1 NeuronLike
Cells (NT2N)
14 Stroke patients (16 y after
stroke)
5 Million cells, 10 million
cells
Improve in: ARAT Seizure Kondziolka
et al
106
Age: 1875 y Intracerebral Cognitive outcome Syncope
Neurological
function
Subdural hematoma
3 SB623 cells (SanBio
Phase I)
Notchtransfected
mesenchymal stromal
cells
18 Stroke patients (660 m after
stroke)
2.5 Million cells, 5 million
cells, 10 million cells
Improve in: Steinberg
et al
108
Age: 1875 y Intracerebral FuglMeyer score
NIHSS > 7 NIHSS
4 SB623 cells (SanBio
Phase II;
ACTISSIMA)
Notchtransfected
mesenchymal stromal
cells
156 patients group 1 (2.5 million
or 5.0 million cells), group 2
(sham placebo); ( > 6<60m
after stroke)
2.5 Million cells, 5 million
cells
Improve in: Wechsler
et al
110
Intracerebral FuglMeyer motor
function total score
ARAT
5 Autologous (ISIS
HERMES trial)
MSCs 31 subacute stroke patients and 11
controls
Intravenous Unpublished Data Detante et al
111
6Allogeneic
(Athersys
MultiStem study)
MAPCs 126 Stroke patients; (48 h) Intravenous Improve in Restricted
employment in
patients with motor
deficits
Hess et al
123,124
FuglMeyer motor
scale
7 CTX0E03 cells
(ReNeuron
PISCES I)
cmycER transgene
human fetal neural stem
cells
Unilateral ischemic stroke
comprising subcortical white
matter or basal ganglia (6 m5y)
2 Million cells, 5 million
cells, 10 million cells, 20
million cells
Improve in: Muir
109
Age: 6085 y Intracerebral NIHSS
NIHSS _6 Ashworth spasticity
Scale
Modest T2 FLAIR
(Continues)
8
|
RIKHTEGAR ET AL.
suppression as a significant mechanism of act of these
cells.
122
After MultiStem administration, there was a
substantial decrease in circulation CD3
+
T cells at
48 hours along with major declines in IL1β,IL6, tumor
necrosis factor (TNFα), and IFNλat 7 days that were
normalized at 30 days.
126
In another recent clinical trial,
developed neurological function was revealed subsequent
implantation of an preserved human neural stemcell line
with no adverse events.
127
Now, ReNeuron, a British
founded company, is sponsoring a Phase I clinical trial,
exploring the clinical use of a CTX0E03; human fetal
neural stem cells genetically altered by insertion of a c
mycER transgene that expresses the cmyc growth factor
when stimulated by 4hydroxytamoxifen, for stroke
treatment.
128
During a Pilot Investigation of Stem Cells
for Stroke (PISCES 1) trial one dose of 2 to 20 million
cells transplanted through stereotaxic intraputaminal
injection to the ipsilesional hemisphere of patients at 6
to 60 months following ischemic stroke and followed for
2 years after treatment.
128,129
In this study, 11 male
patients experienced transplantation with average of 22
months after stroke. Modest developments in motor
function happened in the primary 2 months afterward
transplantation and were sustained subsequently.
109
PISCES 1 informed more milder T2 FLAIR hyperinten-
sities one month after transplantation which was
continued for 12 months.
130
In the PISCES 2 study, 21
male and female patients, 2 to 12 months after stroke
onset, were treated with 20 million CTX0E03 cells
stereotactically through implantation into the putamen.
At 1 year, no safety concerns related to the cells were
reported, and seven patients improved their MRS score
by at least 1 grade. Overall, 15 patients showed
improvement on one or more of the clinical scales.
110
Another doubleblind randomized placebocontrolled
Phase III clinical trial in now in progress which is
investigating the effect of intravenously infusion of
autologous BMderived NSC for patients with stroke
from cerebral infarction.
131
In another study, intracranial
administration of autologous human adult dental pulp
stem cell (DPSC) in patients with chronic stroke was
investigated. The main results were confirmed the safety
and practicability of the treatment; in addition, the
maximum acceptable cell number in humans were
defined.
132
In a new metaanalysis illustrated that stem
cell therapy could considerably improve neurological
functions and quality of life, however, further clinical
trials are required to confirm the clinical use of stem cell
implantation.
133,134
While the most recent clinical trials
using allogeneic cells (SB623, MultiStem, and CTX0E03)
did not need immunosuppression, the potential for
allergic reaction remains. Autologous cells have need of
bone marrow harvest, causing to variable stem cell yield
TABLE 2 (Continued)
Cell type Cell source Patient characteristics
Cell volume and
administration time Outcome Adverse effects Reference
8 CTX0E03 cells
(ReNeuron
PISCES II)
cmyc ER transgene
human fetal neural stem
cells
21 stroke patients; (212 m) 20 Million cells Improve in: Wechsler
et al
110
Intracerebral ARAT
mRS
FuglMeyer
Abbreviations: ARAT, action research arm test; CTX0E03, cmycER transgene human fetal neural stem cells; FLAIR, fluid attenuated inversion recovery; LBS, Layton Bioscience; MAPCs, multipotent adult progenitor
cells; mRS, Modified Rankin Scale (a functional outcome scale with 03 being capable to walk with variable degrees of disability); NIHSS, National Institutes of Health Stroke Scale; NT2N, teratocarcinomaderived
Ntera2/D1 neuronlike cells.
*
(6 m6 y): 6 mo and 6 y after stroke.
RIKHTEGAR ET AL.
|
9
and need time for expansion previous to administration,
nonetheless carry fewer concern for allergic reaction or
rejection.
135
Beside the possible positive consequence of
stem cell therapy, numerous queries have raised on its
clinical applications. Up to date, cell therapies has not yet
reported to be problematical. Affirmation of effectiveness
in randomized, doubleblinded trials are required, how-
ever, many clinical trials are in progress to assess whether
cellbased therapy would develop the subsequent mod-
ality of recovery for stroke consequences. Major clinical
trials investigated stem cells therapies for stroke treat-
ment are summarized in Table 2.
6
|
FUTURE PERSPECTIVES AND
CONCLUSION
Stroke remains the most common reason of death in the
mainstream of developed countries, thus requires serious
attention through conducting preclinical studies at both
acute and chronic stages. Ischemic stroke, prompts acute
neuroinflammation that can aggravate the initial brain
injury.
136
Regenerative medicine in stroke consists of
therapies that could prompt tissue repair leading to
recovery. Numerous alternate attitudes comprising the
usage of ESCs, MSCs, NSC, and iPSCs have been attempted
to treat severe neuronal and functional damages occur
generally after a stroke. Most current clinical trials target to
measure the safety and feasibility of administration routs of
different human adult stem cells in patients with stroke and
aim to define the maximum allowable doses. However,
FDA has not yet accepted any cellbased treatment of acute
and chronic stroke. In summary, cell transplantation for
stroke treatment in humans is still in its infancy. There is a
necessity for more basic and translational studies to
scientifically demonstrate effectiveness of cell therapies in
clinical settings. We anticipate that extra efforts on stem cell
therapy progress, containing clinical trials, will be available
in the immediate prospect.
ACKNOWLEDGMENTS
The authors would like to thank the Aging Research
Institute, Tabriz University of Medical Sciences for
supporting this study.
CONFLICTS OF INTEREST
The authors report no conflicts of interest in this study.
ORCID
Mehdi Yousefi http://orcid.org/0000-0003-0099-6728
REFERENCES
1. Gokcay F, Arsava EM, Baykaner T, et al. Agedependent
susceptibility to infarct growth in women. Stroke. 2011;42:
947951.
2. Hao L, Zou Z, Tian H, Zhang Y, Zhou H, Liu L. Stem cell
based therapies for ischemic stroke. BioMed Res Int. 2014;
2014:20142017.
3. Guzik A, Bushnell C. Stroke epidemiology and risk factor
management. CONTINUUM: Lifelong Learn Neurol. 2017;23:
1539.
4. Saver JL, Goyal M, Van der Lugt A, et al. Time to treatment
with endovascular thrombectomy and outcomes from is-
chemic stroke: a metaanalysis. JAMA. 2016;316:12791289.
5. Reis C, Wilkinson M, Reis H, et al. A look into stem cell
therapy: exploring the options for treatment of ischemic
stroke. Stem Cells Int. 2017;2017:201714.
6. Wang Y, Ji X, Leak RK, Chen F, Cao G. Stem cell therapies in
agerelated neurodegenerative diseases and stroke. Ageing Res
Rev. 2017;34:3950.
7. Dolati S, Ahmadi M, Khalili M, et al. Peripheral Th17/Treg
imbalance in elderly patients with ischemic stroke. Neurol Sci.
2018a;39(4):647654.
8. Zhang Y, Ying G, Ren C, et al. Administration of human
plateletrich plasma reduces infarction volume and improves
motor function in adult rats with focal ischemic stroke. Brain
Res. 2015;1594:267273.
9. Yaghi S, Eisenberger A, Willey JZ. Symptomatic intracerebral
hemorrhage in acute ischemic stroke after thrombolysis with
intravenous recombinant tissue plasminogen activator: a
review of natural history and treatment. JAMA Neurol.
2014;71:11811185.
10. Appireddy RMR, Demchuk AM, Goyal M, et al. Endovascular
therapy for ischemic stroke. J Clin Neurol. 2015;11:18.
11. Del Zoppo GJ, Saver JL, Jauch EC, Adams HP. Expansion of
the time window for treatment of acute ischemic stroke with
intravenous tissue plasminogen activator: a science advisory
from the American Heart Association/American Stroke
Association. Stroke. 2009;40:29452948.
12. Liu X. Beyond the time window of intravenous thrombolysis:
standing by or by stenting? Interv Neurol. 2012;1:315.
13. Dailey T, Metcalf C, Mosley YI, et al. An update on translating
stem cell therapy for stroke from bench to bedside. J Clin Med.
2013;2:220241.
14. Abe K, Yamashita T, Takizawa S, Kuroda S, Kinouchi H,
Kawahara N. Stem cell therapy for cerebral ischemia: from
basic science to clinical applications. J Cerebral Blood Flow
Metab. 2012;32:13171331.
15. Zhang ZG, Chopp M. Neurorestorative therapies for stroke:
underlying mechanisms and translation to the clinic. Lancet
Neurol. 2009;8:491500.
16. Liu X, Ye R, Yan T, et al. Cell based therapies for ischemic
stroke: from basic science to bedside. Prog Neurobiol.
2014;115:92115.
17. Gervois P, Wolfs E, Ratajczak J, et al. Stem cellbased therapies for
ischemic stroke: preclinical results and the potential of imaging
assisted evaluation of donor cell fate and mechanisms of brain
regeneration. Med Res Rev. 2016;36:10801126.
18. Koh SH, Park HH. Neurogenesis in stroke recovery. Transl
Stroke Res. 2017;8:313.
10
|
RIKHTEGAR ET AL.
19. Khoshnam SE, Winlow W, Farzaneh M, Farbood Y,
Moghaddam HF. Pathogenic mechanisms following ischemic
stroke. Neurol Sci. 2017;38:11671186.
20. Azad TD, Veeravagu A, Steinberg GK. Neurorestoration after
stroke. Neurosurg Focus. 2016;40:E2.
21. Bretón RR, Rodríguez JCG 2012. Excitotoxicity and oxidative
stress in acute ischemic stroke. Acute Ischemic Stroke.
InTech.
22. Xu L, Emery JF, Ouyang YB, Voloboueva LA, Giffard RG.
Astrocyte targeted overexpression of Hsp72 or SOD2 reduces
neuronal vulnerability to forebrain ischemia. Glia.
2010;58:10421049.
23. Jung S, Gilgen M, Slotboom J, et al. Factors that determine
penumbral tissue loss in acute ischaemic stroke. Brain.
2013;136:35543560.
24. Iadecola C, Anrather J. The immunology of stroke: from
mechanisms to translation. Nature Med. 2011;17:796808.
25. Pei J, You X, Fu Q. Inflammation in the pathogenesis of
ischemic stroke. Front Biosci (Landmark Ed). 2015;20:772783.
26. Truelsen T, Krarup LH, Iversen HK, et al. Causes of death
data in the global burden of disease estimates for ischemic and
hemorrhagic stroke. Neuroepidemiology. 2015;45:152160.
27. Savitz SI, Cramer SC, Wechsler L, et al. Stem cells as an
emerging paradigm in stroke 3: enhancing the development of
clinical trials. Stroke. 2014;45:634639.
28. Tang YH, Ma YY, Zhang ZJ, Wang YT, Yang GY. Opportu-
nities and challenges: stem cellbased therapy for the
treatment of ischemic stroke. CNS Neurosci Ther. 2015;21:
337347.
29. Chan HH, Wathen CA, Ni M, Zhuo S. Stem cell therapies for
ischemic stroke: current animal models, clinical trials and
biomaterials. RSC Adv. 2017;7:1866818680.
30. Evans JR, Mason SL, Barker RA. Current status of clinical
trials of neural transplantation in Parkinson's disease. Progress
in Brain Research. Elsevier; 2012;200:169198.
31. Stonesifer C, Corey S, Ghanekar S, Diamandis Z, Acosta SA,
Borlongan CV. Stem cell therapy for abrogating stroke
induced neuroinflammation and relevant secondary cell death
mechanisms. Prog Neurobiol. 2017;158:94131.
32. Bang OY. Clinical trials of adult stem cell therapy in patients
with ischemic stroke. J Clin Neurol. 2016;12:1420.
33. Kehat I, KenyaginKarsenti D, Snir M, et al. Human
embryonic stem cells can differentiate into myocytes with
structural and functional properties of cardiomyocytes. J Clin
Invest. 2001;108:407414.
34. Zhang SC, Wernig M, Duncan ID, Brüstle O, Thomson JA. In
vitro differentiation of transplantable neural precursors from
human embryonic stem cells. Nature Biotechnol. 2001;19:
11291133.
35. Stacpoole SRL, Bilican B, Webber DJ, et al. Efficient derivation of
NPCs, spinal motor neurons and midbrain dopaminergic neurons
from hESCs at 3% oxygen. Nat Protoc. 2011;6:12291240.
36. Maroof AM, Keros S, Tyson JA, et al. Directed differentiation
and functional maturation of cortical interneurons from
human embryonic stem cells. Cell Stem Cell. 2013;12:559572.
37. Fujimoto M, Hayashi H, Takagi Y, et al. Transplantation of
telencephalic neural progenitors induced from embryonic
stem cells into subacute phase of focal cerebral ischemia. Lab
Invest. 2012;92:522531.
38. Oyamada N, Itoh H, Sone M, et al. Transplantation of vascular
cells derived from human embryonic stem cells contributes to
vascular regeneration after stroke in mice. J Transl Med.
2008;6:54.
39. Jin K, Xie L, Mao X, et al. Effect of human neural precursor
cell transplantation on endogenous neurogenesis after
focal cerebral ischemia in the rat. Brain Res. 2011;1374:
5662.
40. Zhang P, Li J, Liu Y, et al. Human embryonic neural stem cell
transplantation increases subventricular zone cell prolifera-
tion and promotes periinfarct angiogenesis after focal
cerebral ischemia. Neuropathology. 2011b;31:384391.
41. Kokaia Z, Llorente IL, Carmichael ST. Customized brain cells
for stroke patients using pluripotent stem cells. Stroke.
2018;49:10911098.
42. Zou Z, Zhang Y, Hao L, et al. More insight into mesenchymal
stem cells and their effects inside the body. Expert Opin Biol
Ther. 2010;10:215230.
43. Dolati S, Yousefi M, MahdipourM,etal.Mesenchymalstemcell
and bone marrow mononuclear cell therapy for cardiomyopathy:
from bench to bedside. J Cell Biochem. 2018b;120(1):4555.
44. Fotouhi A, Maleki A, Dolati S, AghebatiMaleki A, Aghebati
Maleki L. Platelet rich plasma, stromal vascular fraction and
autologous conditioned serum in treatment of knee osteoar-
thritis. Biomed Pharmacother. 2018;104:652660.
45. Wakao S, Kuroda Y, Ogura F, Shigemoto T, Dezawa M.
Regenerative effects of mesenchymal stem cells: contribution
of muse cells, a novel pluripotent stem cell type that resides in
mesenchymal cells. Cells. 2012;1:10451060.
46. Bao X, Feng M, Wei J, et al. Transplantation of Flk1 + human
bone marrowderived mesenchymal stem cells promotes
angiogenesis and neurogenesis after cerebral ischemia in rats.
Eur J Neurosci. 2011;34:8798.
47. Tate CC, Fonck C, McGrogan M, Case CC. Human
mesenchymal stromal cells and their derivative, SB623 cells,
rescue neural cells via trophic support following in vitro
ischemia. Cell Transplant. 2010;19:973984.
48. Mitkari B, Kerkelä E, Nystedt J, et al. Intraarterial infusion of
human bone marrowderived mesenchymal stem cells results
in transient localization in the brain after cerebral ischemia in
rats. Exp Neurol. 2013;239:158162.
49. GutiérrezFernández M, RodríguezFrutos B, RamosCejudo J,
et al. Effects of intravenous administration of allogenic bone
marrowand adipose tissuederived mesenchymal stem cells
on functional recovery and brain repair markers in experi-
mental ischemic stroke. Stem Cell Res Ther. 2013;4:11.
50. Honmou O, Houkin K, Matsunaga T, et al. Intravenous
administration of auto serumexpanded autologous mesench-
ymal stem cells in stroke. Brain. 2011;134:17901807.
51. Marei HE, Hasan A, Rizzi R, et al. Potential of stem cellbased
therapy for ischemic stroke. Front Neurol. 2018;9:34.
52. Boese AC, Le QE, Pham D, Hamblin MH, Lee JP. Neural
stem cell therapy for subacute and chronic ischemic stroke.
Stem Cell Res Ther. 2018;9:154.
53. Sinden JD, Vishnubhatla I, Muir KW. Prospects for stem cell
derived therapy in stroke. Progress in Brain Research. Elsevier;
2012;201:119167.
54. Daadi MM, Li Z, Arac A, et al. Molecular and magnetic
resonance imaging of human embryonic stem cellderived
RIKHTEGAR ET AL.
|
11
neural stem cell grafts in ischemic rat brain. Mol Ther.
2009;17:12821291.
55. Goings GE, Sahni V, Szele FG. Migration patterns of
subventricular zone cells in adult mice change after cerebral
cortex injury. Brain Res. 2004;996:213226.
56. Arvidsson A, Collin T, Kirik D, Kokaia Z, Lindvall O.
Neuronal replacement from endogenous precursors in the
adult brain after stroke. Nature Med. 2002;8:963970.
57. Wiltrout C, Lang B, Yan Y, Dempsey R, Vemuganti R.
Repairing brain after stroke: a review on postischemic
neurogenesis. Neurochem Int. 2007;50:10281041.
58. Z Barkho B, Zhao X. Adult neural stem cells: response to
stroke injury and potential for therapeutic applications. Curr
Stem Cell Res Ther. 2011;6:327338.
59. Schouten M, Buijink MR, Lucassen PJ, Fitzsimons CP. New
neurons in aging brains: molecular control by small non
coding RNAs. Front Neurosci. 2012;6:25.
60. Horie N, Pereira MP, Niizuma K, et al. Transplanted stem cell
secreted vascular endothelial growth factor effects poststroke
recovery, inflammation, and vascular repair. Stem Cells.
2011;29:274285.
61. Andres RH, Horie N, Slikker W, et al. Human neural stem
cells enhance structural plasticity and axonal transport in the
ischaemic brain. Brain. 2011;134:17771789.
62. Rikhtegar R, Pezeshkian M, Dolati S, et al. Stem cells as
therapy for heart disease: iPSCs, ESCs, CSCs, and skeletal
myoblasts. Biomed Pharmacother. 2019;109:304313.
63. Oki K, Tatarishvili J, Wood J, et al. Humaninduced
pluripotent stem cells form functional neurons and improve
recovery after grafting in strokedamaged brain. Stem Cells.
2012;30:11201133.
64. Chen SJ, Chang CM, Tsai SK, et al. Functional improve-
ment of focal cerebral ischemia injury by subdural transplan-
tation of induced pluripotent stem cells with fibrin glue. Stem
Cells Dev. 2010b;19:17571767.
65. Kawai H, Yamashita T, Ohta Y, et al. Tridermal tumorigenesis
of induced pluripotent stem cells transplanted in ischemic
brain. J Cerebral Blood Flow Metab. 2010;30:14871493.
66. Yamashita T, Kawai H, Tian F, Ohta Y, Abe K. Tumorigenic
development of induced pluripotent stem cells in ischemic
mouse brain. Cell Transplant. 2011;20:883891.
67. Jiang M, Lv L, Ji H, et al. Induction of pluripotent stem cells
transplantation therapy for ischemic stroke. Mol Cell Biochem.
2011;354:6775.
68. Markus H. Stroke: causes and clinical features. Medicine.
2012;40:484489.
69. Tornero D, Wattananit S, Grønning Madsen M, et al. Human
induced pluripotent stem cellderived cortical neurons inte-
grate in strokeinjured cortex and improve functional recov-
ery. Brain. 2013;136:35613577.
70. Jensen MB, Yan H, KrishnaneyDavison R, Al Sawaf A, Zhang
SC. Survival and differentiation of transplanted neural stem
cells derived from human induced pluripotent stem cells in a
rat stroke model. J Stroke Cerebrovasc Dis. 2013;22:304308.
71. Petit GH, Olsson TT, Brundin P. The future of cell therapies
and brain repair: Parkinson's disease leads the way. Neuro-
pathol Appl Neurobiol. 2014;40:6070.
72. Cyranoski D. Stemcell pioneer banks on future therapies.
Nature. 2012;488:139139.
73. Pietras EM, Warr MR, Passegué E. Cell cycle regulation in
hematopoietic stem cells. J Cell Biol. 2011;195:709720.
74. Felfly H, Muotri A, Yao H, Haddad GG. Hematopoietic stem
cell transplantation protects mice from lethal stroke. Exp
Neurol. 2010;225:284293.
75. Doycheva D, Shih G, Chen H, Applegate R, Zhang JH, Tang J.
Granulocytecolony stimulating factor in combination with
stem cell factor confers greater neuroprotection after hypoxic
ischemic brain damage in the neonatal rats than a solitary
treatment. Transl Stroke Res. 2013;4:171178.
76. Shen LH, Li Y, Chopp M. Astrocytic endogenous glial cell
derived neurotrophic factor production is enhanced by bone
marrow stromal cell transplantation in the ischemic boundary
zone after stroke in adult rats. Glia. 2010;58:10741081.
77. Ponte AL, Marais E, Gallay N, et al. The in vitro migration
capacity of human bone marrow mesenchymal stem cells:
comparison of chemokine and growth factor chemotactic
activities. Stem Cells. 2007;25:17371745.
78. Zacharek A, Chen J, Cui X, et al. Angiopoietin1/Tie2 and
VEGF/Flk1 induced by MSC treatment amplifies angiogenesis
and vascular stabilization after stroke. J Cerebral Blood Flow
Metab. 2007;27:16841691.
79. Chen N, Newcomb J, GarbuzovaDavis S, Sanberg CD,
Sanberg PR, Willing AE. Human umbilical cord blood cells
have trophic effects on young and aging hippocampal neurons
in vitro. Aging Dis. 2010a;1:173.
80. Roura S, Pujal JM, BayesGenis A. Umbilical cord blood for
cardiovascular cell therapy: from promise to fact. Ann NY
Acad Sci. 2012;1254:6670.
81. Chen SH, Wang JJ, Chen CH, et al. Umbilical cord blood
derived CD34 + cells improve outcomes of traumatic brain
injury in rats by stimulating angiogenesis and neurogenesis.
Cell Transplant. 2014b;23:959979.
82. Leonardo CC, Hall AA, Collier LA, Ajmo CT Jr, Jr, Willing
AE, Pennypacker KR. Human umbilical cord blood cell
therapy blocks the morphological change and recruitment of
CD11bexpressing, isolectinbinding proinflammatory cells
after middle cerebral artery occlusion. J Neurosci Res.
2010;88:12131222.
83. Chen C, Wang Y, Yang GY. Stem cellmediated gene
delivering for the treatment of cerebral ischemia: progress
and prospectives. Curr Drug Targets. 2013;14:8189.
84. Zhang L, Li Y, Zhang C, Chopp M, Gosiewska A, Hong K.
Delayed administration of human umbilical tissuederived
cells improved neurological functional recovery in a rodent
model of focal ischemia. Stroke. 2011a;42:14371444.
85. GornickaPawlak E, Janowski M, Habich A, et al. Systemic
treatment of focal brain injury in the rat by human umbilical
cord blood cells being at different level of neural commitment.
Acta Neurobiol Exp (Wars). 2011;71:4664.
86. Sideri A, Neokleous N, De La Grange PB, et al. An overview of
the progress on double umbilical cord blood transplantation.
Haematologica. 2011;2010:0388361220.
87. Fan Y, Shen F, Frenzel T, et al. Endothelial progenitor cell
transplantation improves longterm stroke outcome in mice.
Ann Neurol. 2010;67:488497.
88. Chen C, Lin X, Wang J, et al. Effect of HMGB1 on the
paracrine action of EPC promotes postischemic neovascular-
ization in mice. Stem Cells. 2014a;32:26792689.
12
|
RIKHTEGAR ET AL.
89. Rosell A, Morancho A, NavarroSobrino M, et al. Factors secreted
by endothelial progenitor cells enhance neurorepair responses
after cerebral ischemia in mice. PLoS One. 2013;8:e73244.
90. Chang DJ, Lee N, Park IH, et al. Therapeutic potential of
human induced pluripotent stem cells in experimental stroke.
Cell Transplant. 2013;22:14271440.
91. McGuckin CP, Jurga M, Miller AM, et al. Ischemic brain
injury: a consortium analysis of key factors involved in
mesenchymal stem cellmediated inflammatory reduction.
Arch Biochem Biophys. 2013;534:8897.
92. Savitz SI, Dinsmore J, Wu J, Henderson GV, Stieg P, Caplan
LR. Neurotransplantation of fetal porcine cells in patients
with basal ganglia infarcts: a preliminary safety and feasibility
study. Cerebrovasc Dis. 2005;20:101107.
93. Rabinovich SS, Seledtsov VI, Banul NV, et al. Cell therapy of
brain stroke. Bull Exp Biol Med. 2005;139:126128.
94. RodríguezFrutos B, OteroOrtega L, GutiérrezFernández M,
Fuentes B, RamosCejudo J, DíezTejedor E. Stem cell therapy
and administration routes after stroke. Transl Stroke Res.
2016;7:378387.
95. De Feo D, Merlini A, Laterza C, Martino G. Neural stem cell
transplantation in central nervous system disorders: from cell
replacement to neuroprotection. Curr Opin Neurol. 2012;25:
322333.
96. Mäkelä T, Takalo R, Arvola O, et al. Safety and biodistribu-
tion study of bone marrowderived mesenchymal stromal
cells and mononuclear cells and the impact of the admin-
istration route in an intact porcine model. Cytotherapy. 2015;
17:392402.
97. Misra V, Ritchie MM, Stone LL, Low WC, Janardhan V. Stem
cell therapy in ischemic stroke role of IV and intraarterial
therapy. Neurology. 2012;79:S207S212.
98. Zhang L, Li Y, Romanko M, et al. Different routes of
administration of human umbilical tissuederived cells
improve functional recovery in the rat after focal cerebral
ischemia. Brain Res. 2012;1489:104112.
99. Walczak P, Zhang J, Gilad AA, et al. Dualmodality
monitoring of targeted intraarterial delivery of mesenchymal
stem cells after transient ischemia. Stroke. 2008;39:
15691574.
100. Li L, Jiang Q, Ding G, et al. Effects of administration route on
migration and distribution of neural progenitor cells trans-
planted into rats with focal cerebral ischemia, an MRI study. J
Cerebral Blood Flow Metab. 2010;30:653662.
101. Willing AE, Shahaduzzaman M. Delivery routes for cell therapy in
stroke. Cellbased Therapies in Stroke. Springer; 2013:1528. https://
doi.org/10.1007/978-3-7091-1175-8_2
102. Jiang Y, Wei N, Zhu J, et al. A new approach with less
damage: intranasal delivery of tetracyclineinducible replica-
tiondefective herpes simplex virus type1 vector to brain.
Neuroscience. 2012;201:96104.
103. Liu Z, Li Y, Zhang L, et al. Subacute intranasal administration
of tissue plasminogen activator increases functional recovery
and axonal remodeling after stroke in rats. Neurobiol Dis.
2012;45:804809.
104. Zhong J, Chan A, Morad L, Kornblum HI, Guoping fan U,
Carmichael ST. Hydrogel matrix to support stem cell survival
after brain transplantation in stroke. Neurorehabil Neural
Repair. 2010;24:636644.
105. Kondziolka D, Wechsler L, Goldstein S, et al. Transplantation
of cultured human neuronal cells for patients with stroke.
Neurology. 2000;55:565569.
106. Kondziolka D, Steinberg GK, Wechsler L, et al. Neurotrans-
plantation for patients with subcortical motor stroke: a phase
2 randomized trial. J Neurosurg. 2005;103:3845.
107. Bhasin A, Padma srivastava MV, Mohanty S, Bhatia R,
Kumaran SS, Bose S. Stem cell therapy: a clinical trial of
stroke. Clin Neurol Neurosurg. 2013;115:10031008.
108. Steinberg GK, Kondziolka D, Wechsler LR, et al. Clinical
outcomes of transplanted modified bone marrowderived
mesenchymal stem cells in stroke: a phase 1/2a study. Stroke.
2016;116:0129951824.
109. Muir KW. Clinical trial design for stem cell therapies in stroke:
what have we learned? Neurochem Int. 2017;106:108113.
110. Wechsler LR, Bates D, Stroemer P, AndrewsZwilling YS,
Aizman I. Cell therapy for chronic stroke. Stroke.
2018;49:10661074.
111. Detante O, Jaillard A, Moisan A, et al. Intravenous injection of
autologous mesenchymal stem cells after ischemic stroke
(ISIS/HERMES): protocol and progress. Cerebrovas Dis.
2013;35:852852 Karger Allschwilerstrasse 10, CH4009 Basel,
SWITZERLAND
112. Tsang KS, Ng CPS, Zhu XL, et al. Phase I/II randomized
controlled trial of autologous bone marrowderived mesench-
ymal stem cell therapy for chronic stroke. World J Stem Cells.
2017;9:133.
113. DíezTejedor E, GutiérrezFernández M, MartínezSánchez P,
et al. Reparative therapy for acute ischemic stroke with
allogeneic mesenchymal stem cells from adipose tissue: a
safety assessment: a phase II randomized, doubleblind,
placebocontrolled, singlecenter, pilot clinical trial. J Stroke
Cerebrovasc Dis. 2014;23:26942700.
114. Jiang Y, Zhu W, Zhu J, Wu L, Xu G, Liu X. Feasibility of
delivering mesenchymal stem cells via catheter to the
proximal end of the lesion artery in patients with stroke in
the territory of the middle cerebral artery. Cell Transplant.
2013;22:22912298.
115. Savitz SI, Misra V, Kasam M, et al. Intravenous autologous
bone marrow mononuclear cells for ischemic stroke. Ann
Neurol. 2011;70:5969.
116. Moniche F, Gonzalez A, GonzalezMarcos JR, et al. Intra
arterial bone marrow mononuclear cells in ischemic stroke: a
pilot clinical trial. Stroke. 2012;43:22422244.
117. Steinberg GK, Kondziolka D, Schwartz NE, et al. 2014. A
novel phase 1/2A study of intraparenchymal transplantation
of human modified bone marrow derived cells in patients with
stable ischemic stroke: Am Heart Assoc.
118. Sharma A, Sane H, Gokulchandran N, et al. Autologous bone
marrow mononuclear cells intrathecal transplantation in
chronic stroke. Stroke Res Treat. 2014;2014:20142019.
119. Taguchi A, Sakai C, Soma T, et al. Intravenous autologous
bone marrow mononuclear cell transplantation for stroke:
phase1/2a clinical trial in a homogeneous group of stroke
patients. Stem Cells Dev. 2015;24:22072218.
120. Prasad K, Sharma A, Garg A, et al. Intravenous autologous
bone marrow mononuclear stem cell therapy for ischemic
stroke: a multicentric, randomized trial. Stroke. 2014;
45:36183624.
RIKHTEGAR ET AL.
|
13
121. Moniche F, RosadodeCastro PH, Escudero I, et al. Increasing
dose of autologous bone marrow mononuclear cells trans-
plantation is related to stroke outcome: results from a pooled
analysis of two clinical trials. Stem Cells Int. 2016;2016:
20162018.
122. Yang B, Hamilton JA, Valenzuela KS, et al. Multipotent adult
progenitor cells enhance recovery after stroke by modulating
the immune response from the spleen. Stem Cells. 2017;
35:12901302.
123. Hess D, Clark W, Chiu D, et al. 01. Clinical trial results
acute management results of the doubleblind, rando-
mized, placebo controlled, phase 2 safety and efficacy trial
of Multistem® in adults with ischemic stroke: esoc1152.
Int J Stroke. 2015;10:8.
124. Hess DC, Sila CA, Furlan AJ, Wechsler LR, Switzer JA, Mays
RW. A doubleblind placebocontrolled clinical evaluation of
MultiStem for the treatment of ischemic stroke. Int J Stroke.
2014;9:381386.
125. Bushnell C, Bettger JP, Cockroft KM, et al. Chronic stroke
outcome measures for motor function intervention trials:
expert panel recommendations. Circulation: Cardiovasc Qual-
ity Outcomes. 2015;8:S163S169.
126. Hess DC, Wechsler LR, Clark WM, et al. Safety and efficacy of
multipotent adult progenitor cells in acute ischaemic stroke
(MASTERS): a randomised, doubleblind, placebocontrolled,
phase 2 trial. Lancet Neurol. 2017;16:360368.
127. Sinden JD, Hicks C, Stroemer P, Vishnubhatla I, Corteling R.
Human neural stem cell therapy for chronic ischemic stroke:
charting progress from laboratory to patients. Stem Cells Dev.
2017;26:933947.
128. Kalladka D, Sinden J, Pollock K, et al. Human neural
stemcellsinpatientswithchronic ischaemic stroke
(PISCES): a phase 1, firstinman study. Lancet. 2016;388:
787796.
129. Sinden JD, Muir KW. Stem cells in stroke treatment: the
promise and the challenges. Int J Stroke. 2012;7:426434.
130. George PM, Steinberg GK. Novel stroke therapeutics: un-
raveling stroke pathophysiology and its impact on clinical
treatments. Neuron. 2015;87:297309.
131. Honmou O. Phase III clinical trial using autologous mesench-
ymal stem cells for stroke patients. Nihon rinsho. Japanese J
Clin Med. 2016;74:649654.
132. Nagpal A, Kremer KL, HamiltonBruce MA, et al. TOOTH
(The Open study of dental pulp stem cell Therapy in Humans):
Study protocol for evaluating safety and feasibility of autologous
human adult dental pulp stem cell therapy in patients with
chronic disability after strokeeditor^editors. London, England:
Sage Publications Sage UK; 2016;11(5):575585.
133. Chen L, Zhang G, Khan AA, Guo X, Gu Y. Clinical efficacy
and metaanalysis of stem cell therapies for patients with
brain ischemia. Stem Cells Int. 2016;2016:20162018.
134. Detante O, Moisan A, Hommel M, Jaillard A. Controlled
clinical trials of cell therapy in stroke: metaanalysis at six
months after treatment. International Journal of Stroke.
2017;12:748751.
135. Kenmuir CL, Wechsler LR. Update on cell therapy for stroke.
Stroke Vasc Neurol. 2017;2:5964.
136. Jin R, Liu L, Zhang S, Nanda A, Li G. Role of inflammation
and its mediators in acute ischemic stroke. J Cardiovasc Trans
Res. 2013;6:834851.
How to cite this article: Rikhtegar R, Yousefi M,
Dolati S, et al. Stem cellbased cell therapy for
neuroprotection in stroke: A review. J Cell Biochem.
2018;114. https://doi.org/10.1002/jcb.28207
14
|
RIKHTEGAR ET AL.
... ESC-derived mesenchymal stem cells, vascular progenitor cells, and neural progenitor cells have shown beneficial effects without evidence of tumorigenesis [28]. Neuronal progenitor cells derived from ESCs can reduce infarct volume, promote neurogenesis, and enhance functional recovery [29]. Transplanted embryonic neural stem cells have been shown to stimulate the release of angiogenic cytokines, leading to vascular endothelial proliferation within 15 day post-cerebral ischemia [29]. ...
... Neuronal progenitor cells derived from ESCs can reduce infarct volume, promote neurogenesis, and enhance functional recovery [29]. Transplanted embryonic neural stem cells have been shown to stimulate the release of angiogenic cytokines, leading to vascular endothelial proliferation within 15 day post-cerebral ischemia [29]. ...
Article
Full-text available
This paper explores the potential of stem cell therapies in revolutionising stroke recovery, addressing the limitations of current treatments and emphasising regenerative medicine as a promising alternative. Stroke, a leading cause of disability and death worldwide, necessitates innovative approaches due to the temporal constraints and regen-erative deficiencies in existing therapeutic modalities. The review explores the diverse mechanisms underlying stem cell-mediated recovery, encompassing neuroprotection, neurogenesis, angiogenesis, modulation of inflammatory responses, and induction of host brain plasticity. We searched prominent databases (PubMed, Scopus, Google Scholar, and Web of Science) from inception to January 2024 for studies on "stem cell therapy" or "regenerative medicine" combined with "stroke recovery" or "cerebrovascular accident". Studies in humans and animals, published in peer-reviewed journals, and investigating the impact of stem cell therapy on stroke recovery were included. We excluded non-English publications and those lacking sufficient outcome data. Evidence from animal studies demonstrates the efficacy of various stem cell types, while human studies, though limited, contribute valuable insights into safety and potential efficacy. Safety considerations, crucial for successful clinical application, emphasise the need for rigorous preclinical and clinical studies, long-term follow-up data, and ethical standards. Challenges in the field, such as study design heterogeneity, optimising stem cell delivery methods, and identifying subpopulations likely to benefit, require concerted efforts to overcome. Standardising methodologies, refining delivery routes, and personalising interventions based on biomarkers are essential. This review positions stem cell therapies as promising for comprehensive neural tissue recovery following stroke.
... New therapeutic agents and techniques should have ability to amplify endogenous processes for tissue regeneration such as angiogenesis, neurogenesis, and axonal outgrowth [18]. Stem cell based therapy has been known as a promising strategy to attenuate neurological disorders such as cerebral ischemia/reperfusion injury and improve life quality of patients [19][20][21][22][23][24]. Mesenchymal stem cells (MSCs) have attracted tremendous interest in treatment of ischemic diseases such as stroke owing to their unique properties such as ability for modulation of the inflammatory responses, low immunogenicity, easy attainability, and differentiation plasticity [25][26][27][28][29][30][31]. ...
... Later, researchers found that neuroprotective effects of grafted MSCs are associated with promoting angiogenesis, neurogenesis and axonal growth through targeting different cellular pathways [36,57]. Although some researchers have shown that stem cell therapy of ischemic therapy is safe, grafting cells might have some complications such as undesirable immune responses and tumor induction [23,58]. To tackle the limitations of cell therapy of ischemic stroke, the researchers have focused on paracrine effects of stem cells [48]. ...
Article
Full-text available
It has been reported that the therapeutic potential of stem cells is mainly mediated by their paracrine factors. In order to identify the effects of conditioned medium of mesenchymal stem cells (MSC-CM) against stroke, a systematic review was conducted. We searched PubMed, Scopus, and ISI Web of Science databases for all available articles relevant to the effects of MSC-CM against the middle cerebral artery occlusion (MCAO) model of ischemic stroke until August 2022. The quality of the included studies was evaluated using The STAIR scale. During the systematic search, a total of 356 published articles were found. A total of 15 datasets were included following screening for eligibility. The type of cerebral ischemia was the MCAO model and CM was obtained from MSCs. The results showed that the therapeutic time window can be considered a crucial factor when researchers use MSC-CM for stroke therapy. In addition, MSC-CM therapy contributes to functional recovery and reduces infarct volume after stroke by targeting different cellular signaling pathways. Our findings showed that MSC-CM therapy has the ability to improve functional recovery and attenuate brain infarct volume after ischemic stroke in preclinical studies. We hope our study accelerates needed progress towards clinical trials.
... Acute ischemic stroke (AIS), myocarditis, and cancer are leading reasons for mortality in developing and industrialized nations, particularly among the elderly (Rikhtegar et al., 2019). Effective management of diseases is challenging and lengthy. ...
... Numerous studies have proved the distinctive performance of biomedical cell-loaded scaffolds for tissue regeneration [10]. Cultured cells particularly stem cells, with their characteristic capacity to differentiate into various cell types, have shown significant benefits in in dermal wound healing [11]. ...
Article
Full-text available
Burn wounds are the most destructive and complicated type of skin or underlying soft tissue injury that are exacerbated by a prolonged inflammatory response. Several cell-based therapeutic systems through the culturing of potent stem cells on modified scaffolds have been developed to direct the burn healing challenges. In this context, a new regenerative platform based on boron (B) enriched-acellular sheep small intestine submucosa (AOSIS) scaffold was designed and used as a carrier for mesenchymal stem cells derived from Wharton's jelly (WJMSCs) aiming to promote the tissue healing in burn-induced rat models. hWJMSCs have been extracted from human extra-embryonic umbilical cord tissue. Thereafter, 96 third‐degree burned Wistar male rats were divided into 4 groups. The animals that did not receive any treatment were considered as group A (control). Then, group B was treated just by AOSIS scaffold, group C was received cell-seeded AOSIS scaffold (hWJMSCs-AOSIS), and group D was covered by boron enriched-cell-AOSIS scaffold (B/hWJMSCs-AOSIS). Inflammatory factors, histopathological parameters, and the expression levels of epitheliogenic and angiogenic proteins were assessed on 5, 14 and 21 days post-wounding. Application of the B/hWJMSCs-AOSIS on full-thickness skin-burned wounds significantly reduced the volume of neutrophils and lymphocytes at day 21 post-burning, whilst the number of fibroblasts and blood vessels enhanced at this time. In addition, molecular and histological analysis of wounds over time further verified that the addition of boron promoted wound healing, with decreased inflammatory factors, stimulated vascularization, accelerated re-epithelialization, and enhanced expression levels of epitheliogenic genes. In addition, the boron incorporation amplified wound closure via increasing collagen deposition and fibroblast volume and activity. Therefore, this newly fabricated hWJMSCs/B-loaded scaffold can be used as a promising system to accelerate burn wound reconstruction through inflammatory regulation and angiogenesis stimulation.
... Most IS has a sudden onset followed by a rapid progression; thus, the limited expansion of Treg cells at the beginning cannot meet the need for conducting immunosuppressive effects over time. Human umbilical cord blood (UCB) has become a commonly used source for cell therapy, and the relative scarcity of CD25 + non-Treg cells in UCB makes the purification of nTreg cells from UCB more convenient compared to isolating them from peripheral blood (Rikhtegar et al., 2019;Xia et al., 2016). Implanting lymphocytes that were co-cultivated with human UCB stem cells into the peripheral blood stream resulted in the amelioration of neurological deficits, a reduction in the volume of tissue damage from infarction, and the dampening of neuronal apoptosis. ...
... Within minutes of a stroke, millions of brain neurons perish (Saver, 2006;Overgaard, 2014). Although stroke-related cell death is irreversible, stem cell therapy may be of assistance (Wang et al., 2012;Rikhtegar et al., 2019). In light of this, Singh et al. conducted They put a strong emphasis on making the therapy translational. ...
... Stroke is a neurological disorder that is divided into ischemic and hemorrhagic types [4,5]. In the ischemic type, the hypoxia of the brain tissue occurs with the cessation of blood flow to the brain tissue and leads to the destruction of neurons and glial cells [2,3,6,7]. The sequence of the events responding to ischemia is known as the ischemic cascade, including glutamate release, calcium influx, OS, inflammation, and ultimately, apoptosis, which leads to irreversible neuronal death [8,9]. ...
Article
Full-text available
Methods: Sixty patients with a mean age of 68.60 ± 2.10 comprising 29 females (48.33%), who were admitted to an academic tertiary care facility within the first 12 hours poststroke symptoms onset or last known well (LKW), in case symptom onset time is not clear, were included in this study. AIS was confirmed based on a noncontrast head CT scan and also neurological symptoms. Patients were randomly and blindly assigned to OEA of 300 mg/day (n = 20) or 600 mg/day (n = 20) or placebo (n = 20) in addition to the standard AIS treatment for three days. A blood sample was drawn at 12 hours from symptoms onset or LKW as the baseline followed by the second blood sample at 72 hours post symptoms onset or LKW. Blood samples were assessed for inflammatory and biochemical parameters, oxidative stress (OS) biomarkers, and lipid profile. Results: Compared to the baseline, there is a significant reduction in the urea, creatinine, triglyceride, high-density lipoprotein, cholesterol, alanine transaminase, total antioxidant capacity, malondialdehyde (MDA), total thiol groups (TTG), interleukin-6 (IL-6), and C-reactive protein levels on the follow-up blood testing in the OEA (300 mg/day) group. In patients receiving OEA (600 mg/day) treatment, there was only a significant reduction in the MDA level comparing baseline with follow-up blood testing. Also, the between-group analysis revealed a statistically significant difference between patients receiving OEA (300 mg/day) and placebo in terms of IL-6 and TTG level reduction when comparing them between baseline and follow-up blood testing. Conclusion: OEA in moderate dosage, 300 mg/day, add-on to the standard stroke treatment improves short-term inflammatory, OS, lipid, and biochemical parameters in patients with AIS. This effect might lead to a better long-term neurological prognosis.
Article
Full-text available
A baixa capacidade regenerativa do cérebro humano representa um grande desafio no tratamento de doenças neurológicas e neurodegenerativas. Embora essas doenças apresentem alta morbidade e mortalidade, os tratamentos existentes são pouco eficazes para reverter o quadro, muitas vezes, servindo apenas como forma de alívio dos sintomas. A regeneração é o processo de reparo de células, tecidos ou órgãos que passaram por algum processo de dano, e têm o objetivo de restaurar as funções vitais desempenhadas por aquela região. As células-tronco são células indiferenciadas com grande capacidade proliferativa e apresentam protagonismo nesse processo. As células-tronco são classificadas de acordo com a sua capacidade de diferenciação, e podem ser separadas em totipotentes ou onipotentes, pluripotentes, multipotentes, oligopotentes e unipotentes. Considerando a necessidade crescente de tratamentos alternativos para danos ao cérebro e o papel primordial das células-tronco no processo regenerativo, a utilização dessas células para a recuperação de áreas cerebrais danificadas vem crescendo exponencialmente no campo da medicina regenerativa. Já existem atualmente testes clínicos para a aplicação dessa tecnologia para o tratamento de doenças neurodegenerativas como a Doença de Parkinson e a Doença de Alzheimer. Além disso, essa ferramenta também vem sendo testada para o tratamento de danos cerebrais causados por condições como Acidente Vascular Cerebral (AVC), traumatismo craniano, e, até mesmo, para reverter os efeitos colaterais da quimioterapia. Embora essa tecnologia apresente grande potencial, ela ainda se encontra na fase de desenvolvimento, sendo necessária a realização de um maior número de testes para que seus mecanismos possam ser esclarecidos e para que a sua segurança e eficácia possa ser comprovada. Ressaltamos ainda a importância de criar uma infraestrutura urbana propícia ao avanço da medicina regenerativa, destacando a colaboração entre instituições de pesquisa, hospitais e empresas de biotecnologia. Ao examinar políticas públicas que promovem a pesquisa em células-tronco, enfatizamos como o planejamento urbano pode transformar cidades em centros de excelência, atraindo investimentos, talentos e promovendo o desenvolvimento econômico
Article
Cerebral ischemic damage is prevalent and the second highest cause of death globally across patient populations; it is as a substantial reason of morbidity and mortality. Mesenchymal stromal cells (MSCs) have garnered significant interest as a potential treatment for cerebral ischemic damage, as shown in ischemic stroke, because of their potent intrinsic features, which include self-regeneration, immunomodulation, and multi-potency. Additionally, MSCs are easily obtained, isolated, and cultured. Despite this, there are a number of obstacles that hinder the effectiveness of MSC-based treatment, such as adverse microenvironmental conditions both in vivo and in vitro. To overcome these obstacles, the naïve MSC has undergone a number of modification processes to enhance its innate therapeutic qualities. Genetic modification and preconditioning modification (with medications, growth factors, and other substances) are the two main categories into which these modification techniques can be separated. This field has advanced significantly and is still attracting attention and innovation. We examine these cutting-edge methods for preserving and even improving the natural biological functions and therapeutic potential of MSCs in relation to adhesion, migration, homing to the target site, survival, and delayed premature senescence. We address the use of genetically altered MSC in stroke-induced damage. Future strategies for improving the therapeutic result and addressing the difficulties associated with MSC modification are also discussed.
Article
Full-text available
Stem cell therapy, which has promising results in acute disorders such as stroke, supports treatment by providing rehabilitation in the chronic stage patients. In acute stroke, thrombolytic medical treatment protocols are clearly defined in neurologic emergencies, but in neurologic patients who miss the “thrombolytic treatment intervention window,” or in cases of hypoxic-ischemic encephalopathy, our hands are tied, and we are still unfortunately faced with hopeless clinical implementations. We consider mesenchymal stem cell therapy a viable option in these cases. In recent years, novel research has focused on neuro-stimulants and supportive and combined therapies for stroke. Currently, available treatment options are limited, and only certain patients are eligible for acute treatment. In the scope of our experience, five stroke patients were evaluated in this study, who was treated with a single dose of 1–2 × 10 ⁶ cells/kg allogenic umbilical cord-mesenchymal stem cells (UC-MSCs) with the official confirmation of the Turkish Ministry of Health Stem Cell Commission. The patients were followed up for 12 months, and clinical outcomes are recorded. NIH Stroke Scale/Scores (NIHSS) decreased significantly ( p = 0.0310), and the Rivermead Assessment Scale (RMA) increased significantly ( p = 0.0234) for all patients at the end of the follow-up. All the patients were followed up for 1 year within a rehabilitation program. Major clinical outcome improvements were observed in the overall clinical conditions of the UC-MSC treatment patients. We observed improvement in the patients’ upper extremity and muscle strength, spasticity, and fine motor functions. Considering recent studies in the literature together with our results, allogenic stem cell therapies are introduced as promising novel therapies in terms of their encouraging effects on physiological motor outcomes.
Article
Full-text available
Background: The alarming disability burden and a high prevalence rate of stroke in India has encouraged the researchers to develop regenerative therapies to reduce clinical deficits. This study evaluates safety, feasibility and efficacy of autologous mononuclear and mesenchymal cell transplantation in stroke patients evaluated on clinical scores and functional imaging (fMRI and DTI). Methods: Forty (n=40) stroke patients were recruited with the inclusion criteria as: 3 months to 2 years of index event, power of hand muscles of at least 2; Brunnstrom stage: 2-5; conscious and comprehendible. Fugl Meyer (FM), modified Barthel Index (mBI), Medical Research Council (MRC) grade for strength, Ashworth tone scale and functional imaging was used for assessments at baseline, 8 weeks and 24 weeks. 50-60 million cells in 250 ml saline were infused intravenously over 2-3 h. Results: The safety test profile was normal with no mortality or cell related adverse reactions in stem cell patients. Among outcome parameters, only modified Barthel Index (mBI) showed statistical significant improvement (p<0.05) in the stem cell group. An increased number of cluster activation in Brodmann areas BA 4, BA 6 was observed post stem cell infusion indicating neural plasticity. Conclusion: Autologous intravenous stem cell therapy is safe and feasible. Stem cells act as "scaffolds" for neural transplantation and may aid in repair mechanisms in stroke.
Article
Full-text available
Neural stem cells (NSCs) play vital roles in brain homeostasis and exhibit a broad repertoire of potentially therapeutic actions following neurovascular injury. One such injury is stroke, a worldwide leading cause of death and disability. Clinically, extensive injury from ischemic stroke results from ischemia-reperfusion (IR), which is accompanied by inflammation, blood-brain barrier (BBB) damage, neural cell death, and extensive tissue loss. Tissue plasminogen activator (tPA) is still the only US Food and Drug Administration–approved clot-lysing agent. Whereas the thrombolytic role of tPA within the vasculature is beneficial, the effects of tPA (in a non-thrombolytic role) within the brain parenchyma have been reported as harmful. Thus, new therapies are needed to reduce the deleterious side effects of tPA and quickly facilitate vascular repair following stroke. The Stroke Treatment Academic Industry Roundtable (STAIR) recommends that stroke therapies “focus on drugs/devices/treatments with multiple mechanisms of action and that target multiple pathways”. Thus, based on multifactorial ischemic cascades in various stroke stages, effective stroke therapies need to focus on targeting and ameliorating early IR injury as well as facilitating angiogenesis, neurogenesis, and neurorestorative mechanisms following stroke. This review will discuss the preclinical perspectives of NSC transplantation as a promising treatment for neurovascular injury and will emphasize both the subacute and chronic phase of ischemic stroke.
Article
Full-text available
Ischemic stroke is one of the major health problems worldwide. The only FDA approved anti-thrombotic drug for acute ischemic stroke is the tissue plasminogen activator. Several studies have been devoted to assessing the therapeutic potential of different types of stem cells such as neural stem cells (NSCs), mesenchymal stem cells, embryonic stem cells, and human induced pluripotent stem cell-derived NSCs as treatments for ischemic stroke. The results of these studies are intriguing but many of them have presented conflicting results. Additionally, the mechanism(s) by which engrafted stem/progenitor cells exert their actions are to a large extent unknown. In this review, we will provide a synopsis of different preclinical and clinical studies related to the use of stem cell-based stroke therapy, and explore possible beneficial/detrimental outcomes associated with the use of different types of stem cells. Due to limited/short time window implemented in most of the recorded clinical trials about the use of stem cells as potential therapeutic intervention for stroke, further clinical trials evaluating the efficacy of the intervention in a longer time window after cellular engraftments are still needed.
Article
Full-text available
Neural stem cells (NSCs) offer a potential therapeutic benefit in the recovery from ischemic stroke. Understanding the role of endogenous neural stem and progenitor cells under normal physiological conditions aids in analyzing their effects after ischemic injury, including their impact on functional recovery and neurogenesis at the site of injury. Recent animal studies have utilized unique subsets of exogenous and endogenous stem cells as well as preconditioning with pharmacologic agents to better understand the best situation for stem cell proliferation, migration, and differentiation. These stem cell therapies provide a promising effect on stimulation of endogenous neurogenesis, neuroprotection, anti-inflammatory effects, and improved cell survival rates. Clinical trials performed using various stem cell types show promising results to their safety and effectiveness on reducing the effects of ischemic stroke in humans. Another important aspect of stem cell therapy discussed in this review is tracking endogenous and exogenous NSCs with magnetic resonance imaging. This review explores the pathophysiology of NSCs on ischemic stroke, stem cell therapy studies and their effects on neurogenesis, the most recent clinical trials, and techniques to track and monitor the progress of endogenous and exogenous stem cells
Article
Heart Diseases are serious and global public health concern. In spite of remarkable therapeutic developments, the prediction of patients with Heart Failure (HF) is weak, and present therapeutic attitudes do not report the fundamental problem of the cardiac tissue loss. Innovative therapies are required to reduce mortality and limit or abolish the necessity for cardiac transplantation. Stem cell–based therapies applied to the treatment of heart disease is according to the understanding that natural self-renewing procedures are inherent to the myocardium, nonetheless may not be adequate to recover the infarcted heart muscle. Following the first account of cell therapy in heart diseases, examination has kept up to rapidity; besides, several animals and human clinical trials have been conducted to preserve the capacity of numerous stem cell population in advance cardiac function and decrease infarct size. The purpose of this study was to censoriously evaluate the works performed regarding the usage of four major subgroups of stem cells, including induced Pluripotent Stem Cells (iPSC), Embryonic Stem Cells (ESCs), Cardiac Stem Cells (CDC), and Skeletal Myoblasts, in heart diseases, at the preclinical and clinical studies. Moreover, it is aimed to argue the existing disagreements, unsolved problems, and prospect directions.
Article
To date, stem cell–based therapies for cardiac diseases have not achieved any significant clinical accomplishment. Globally, numerous patients are currently treated with autologous stem cells. The safety and practicality of this technique have been well‐examined, its disadvantages have been recognized, and many trials have been proposed. Inadequate description of the implemented cell types, a variety of cell‐handling proficiencies, and concerning factors related to autologous stem cells have been known as the central elements restricting the approval of cell‐based therapies. The idea that bone marrow (BM)‐derived cells could be applied to regenerate and cure damage in various organs is the basis for bone marrow mononuclear cell (BMMNC) therapy for heart disease. Mesenchymal stem cells (MSCs) are a part of the BMMNCs; on one hand, they have the capability to differentiate into various tissues, and, on the other, their immunomodulatory effects have been considered and clinically confirmed in different experiments. In this review, we summarize the knowledge obtained by trials in which mesenchymal cell–based therapy has been practiced. Furthermore, we accentuate the developments in the purification and lineage specification of MSCs as well as BMMNCs that have influenced the progress of future stem cell–based therapies with special attention on cardiovascular disease.
Article
Osteoarthritis (OA) is a multifactorial chronic disease, causing several problems on patients, hygiene and community care systems. Conventional therapies, such as non-pharmacological mediations, systemic drug treatment and intra-articular therapies are applying previously; however, controlling and management approaches of the disease mainly remain insufficient. Injections of intra-articular therapies directly into the joint evade conservative obstacles to joint entry, rise bioavailability and minor systemic toxicity. Current progresses in osteoarthritis management have designed better diversity of treatment approaches. Innovative treatments, such as autologous blood products and mesenchymal stem cells, are in progress. Platelet-rich plasma (PRP) is one of the several novel therapeutic approaches that stay to progress in the field of orthopedic medicine. Stromal vascular fraction (SVF) comprises a lesser amount of mesenchymal stem cells and is a treatment for OA and cartilage damage. Based on novel opinions, an innovative therapy by autologous conditioned serum (ACS) from the whole blood was settled. The inoculation of ACS into tissues has revealed clinical efficacy for the treatment of osteoarthritis and muscle injuries. Here, we make available historical perspective of PRP, SVF, and ACS and the other existing researches on using PRP, SVF and ACS for the treatment of knee OA. In conclusion, in current years, OA stem cell therapy has rapidly progressed, with optimistic consequences in animals and human studies. Additionally, PRP, SVF and ASC injection seem to be accompanied with numerous favorable results for treatment of patients with OA.
Article
CD4⁺CD25⁺ regulatory T (Treg) cells and Th17 cells play important roles in peripheral immunity. Immune responses are main elements in the pathogenesis of ischemic stroke (IS). The contribution of Th17 cells in IS patients has not been proved, and whether the balance of Treg/Th17 cells is changed in IS patients remains unidentified. In the present study, we studied Th17 and Treg cell frequency, cytokine secretion, expression of transcription factors, and microRNAs related to Th17 and Treg cells differentiation, which is compared between IS patients and control group. Thirty patients with IS and 30 individuals as control group were enrolled in this study. The frequency of Th17 and Treg lymphocytes, the expression of transcription factors and microRNAs related to these cells, and the serum levels of associated cytokines were assessed by flow cytometry, real-time PCR, and ELISA, respectively. A significant reduction in proportion of peripheral Treg cell frequency and the levels of TGF- 훽 and FOXP3 expression were observed in patients with IS compared with controls, while the proportions of Th17 were increased dramatically, and these effects were along with increases in the levels of IL-17A and RORγt expression in IS patients. The levels of mir-326 and mir-106b-25 expression were increased in patients with IS. These studies suggest that the increase in proportion of Th17 cells and decrease in Treg cells might contribute to the pathogenesis of IS. Manipulating the balance between Tregs and Th17 cells might be helpful for the treatment of IS.