Mesenchymal stem cells in the treatment of ischemic stroke

Cover Page


Cite item

Abstract

Over the past two decades, multiple preclinical studies have shown that transplantation of mesenchymal stem cells leads to a pronounced positive effect in animals with experimental stroke. Based on the promising results of preclinical studies, several clinical trials on the transplantation of mesenchymal stem cells to stroke patients have also been conducted. In this review, we present and analyze the results of completed clinical trials dedicated to the mesenchymal stem cells transplantation in patients with ischemic stroke. According to the obtained results, it can be concluded that transplantation of mesenchymal stem cells is safe and feasible from the economic and biomedical point of view. For the further implementa-tion of this promising approach into the clinical practice, randomized, placebo-controlled, multicenter clinical trials are needed with a large sample of patients and optimized cell transplantation protocols and patient inclusion criteria. In this review we also discuss possi-ble strategies to enhance the effectiveness of cell therapy with the use of mesenchymal stem cells.

Full Text

INTRODUCTION

Ischemic stroke (IS) remains a crucial issue in modern medicine owing to its high morbidity, mortality, and patient disability rates [1–3]. According to the World Health Organization, IS and other acute circulatory disorders account for 11% of global mortality [4–6]. Over the last 5 years, ~430,000–470,000 cases of infectious diseases have been reported annually in Russia. The hospital mortality rate ranged from 17% to 21% [7].

Currently, the only effective IS treatments are intravenous thrombolysis and intravascular thromboextraction during the acute period to restore blood flow in cerebral arteries [8, 9]. However, these methods have limitations and contraindications, the most significant of which is the short time interval or “therapeutic window” for their application. Recent studies have shown that the therapeutic window ranges from 4.5 h for intravenous thrombolysis to 24 h for thromboextraction [10–12]. After IS, patients may experience lifelong neurological deficits, even after successful reperfusion therapy, due to the death of neurons and glial cells in the center of the brain infarction [13].

Recent analytical studies have revealed that in Russia, a high percentage of patients become permanently disabled after experiencing IS, resulting in reduced ability to work and contributing to economic cost. The state incurs an average cost of 0.9–1.2 million rubles per stroke case, and the economic damage during the first year after a stroke is equivalent to 0.3% of the country’s annual gross domestic product. This highlights the creation of a substantial socioeconomic burden for the state and thus confirms the need for new, effective methods and approaches for IS therapy in patients who miss the therapeutic window.

In recent years, there has been significant evidence supporting the potential of mesenchymal stem cell (MSC) transplantation as an IS therapy. MSCs are a subpopulation of mesenchymal stromal cells that meet the stemness criteria established by the International Society for Cellular Therapy (ISCT) [14]. The stemness criteria imply cell multipotency, i.e., the ability to differentiate into different mature cell types within one germinal leaflet (in the case of MSCs, mesoderm) along with the ability to actively proliferate. MSCs are commonly used in cell therapy and regenerative medicine because of their immunomodulatory, anti-inflammatory, angiogenesis-stimulating, and antiapoptotic effects [15, 16]. In addition to directly secreting cytokines and other regulatory molecules into the extracellular space, MSCs produce extracellular vesicles that can be taken up by target cells, facilitating the intracellular delivery of biologically active molecules. Exosomes are the most extensively studied MSC extracellular vesicles. They are 30–100 nm in size and can transport a diverse range of regulatory microRNAs and growth factors.

Phenotypically, MSCs express certain cell surface markers from the differentiation cluster (CD). The ISCT Mesenchymal and Tissue Stem Cell Committee proposed the minimum criteria for defining MSCs as cells that express CD105, CD73, and CD90 but do not express CD45, CD34, CD14, CD11b, CD79α, CD19, or HLA-DR. As adherent and actively proliferating cells, they can form a dense cell monolayer when cultured on adhesive polymer coatings under standard conditions. Additionally, under special conditions in vitro, MSCs can differentiate into adipocytes, chondroblasts, and osteoblasts [17] and into various cell types, such as pericytes, hepatocytes [18], cardiomyocytes [19], and other mesodermal cells. However, the scientific community currently questions the ability of MSCs to differentiate into neurons without additional induction [20].

MSCs can be isolated from various organs and tissues, including the bone marrow, placenta, umbilical cord, amniotic fluid, adipose tissue, skin, dental pulp, and stroma of parenchymal organs [21–26]. Importantly, MSCs obtained from different sources or donors may exhibit significant differences in gene expression patterns, resulting in varying regenerative potential [27]. MSCs have an immunomodulatory effect and are considered relatively immunologically inert because of their low HLA expression. This means that the risk of pathological immunosensitization during allogeneic MSC transplantation is low although it cannot be excluded [28].

The advantages of MSCs include their ease to procure from primary sources, such as the placenta, which is classified as biowaste, and the low cost of obtaining primary cultures and scaling up cell production for clinical use. Furthermore, their safety has been confirmed in preclinical and clinical studies. Throughout the period of MSC research, no cases of tumor formation or oncogenic transformation resulting from transplanted MSCs have been reported. Therefore, the oncological safety of these cells is not debatable [29, 30].

Numerous preclinical studies (PCSs) have found that transplantation of MSCs in animals with experimental brain infarction can have a significant positive therapeutic effect [31–33]. Studies have demonstrated that systemic (intravenous or intra-arterial) MSC transplantation and local intracerebral injection improve the survival rate of laboratory animals, reduce the severity of neurological deficits, and, in some cases, decrease the size of the brain infarction zone [34–40]. According to PCS data, the most favorable MSC transplantation method in IS, particularly in the acute and subacute phases, is systemic intra-arterial administration. This method allows MSCs to enter the brain microcirculatory bed first and potentially have the greatest systemic effect within the central nervous system [34, 41]. However, the optimal method of MSC transplantation remains unclear. Intravenous administration is the least invasive method of systemic administration. However, most cells are retained in parenchymal organs, primarily the lungs, liver, and spleen. This retention reduces the efficiency of MSC delivery to the brain, resulting in lower functional recovery compared with intra-arterial administration [42]. Intra-arterial transplantation has shown the highest therapeutic efficacy by delivering MSCs directly to cerebral arteries, bypassing peripheral organs [43]. To avoid embolic complications during intra-arterial injection, an appropriate dose and rate of MSC administration should be selected [44]. In recent years, endovascular surgery techniques have undergone significant development, making intra-arterial access more readily available for routine clinical use.

The ideal timing for transplantation of MSCs and other types of stem cells in stroke patients remains unknown. However, unlike reperfusion therapy, the therapeutic window for cell therapy in IS is longer. The therapeutic effects of cell transplantation from several hours to several months after the onset of acute cerebral circulatory failure have been reported [40]. In comparing the therapeutic effects, the experimental animals showed the best functional recovery and faster reduction of the brain infarction center volume within 24–48 h from the onset of neurological symptoms [45, 46]. The mechanism of action of MSCs is associated with their anti-inflammatory and neuroprotective effects in brain ischemia, neuroinflammation, and blood–brain barrier damage. This association determines the maximum efficacy of MSC therapy within the first 48 h after IS [47]. The unique immunological properties of MSCs enable their allogeneic transplantation, which is significant from a socioeconomic perspective. This allows for the mass production of placental MSCs and their use during the acute and acute phases of IS when autologous cells are unobtainable [46, 48, 49].

The properties of MSCs described above and the encouraging results of PCSs have made it possible to conduct the first clinical trials (CTs) in foreign countries to study the effect of MSC transplantation on the course and outcomes of IS in humans.

The present study aimed to summarize the clinical trial experience of MSC transplantation in CTs, present the modern concept of MSC mechanism of action based on evidence-based medicine, and outline the methods of developing cell therapy for IS.

CLINICAL TRIAL RESULTS

Currently, the scientific literature presents the results of 14 CTs. These trials were conducted in foreign countries and evaluated the safety and efficacy of transplantation of MSCs obtained from different sources. Patients were transplanted with both autologous and allogeneic MSCs and genetically modified MSCs. In all studies, patients with IS were treated with standard therapy according to the clinical recommendations accepted in the country, in addition to cell therapy. Table 1 presents the results of the conducted studies [50–63].

 

Table 1

Results of major clinical trials on cell therapy for ischemic stroke

Study

Phase

Total patients/MSC transplantation, n

MSC

type/dose

Administration route

Follow-up period

Efficacy/adverse events,

if identified

Bang et al. 2005 [50]

I/II

30/5

Autologous (bone marrow)/two doses of 50 million cells

Intravenous

1 year

NIHSS

improvement

Lee et al. [51]

I/II

52/16

Autologous (bone marrow)

Intravenous transplantation

5 years

Improved functional recovery, reduced mortality

Honmou

et al. [52]

I

12

Autologous MSCs (bone marrow)/0.6 to 1.6×108 cells

Intravenous transplantation

1 year

Trend towards reduction in neurological deficits, significant reduction in size of infarct focus

Bhasin

et al. [53]

I/II

12/6

Autologous MSCs (bone marrow) /

50–60×106 cells

Intravenous transplantation

6 months

Trend towards reduction in neurological

deficits

Bhasin

et al. [54]

I/II

40/ MSC 6/

14 mononuclear SCs

Autologous MSCs (bone marrow)/

50–60 million cells

Intravenous transplantation

6 months

Statistically significant improvement

of mBI in the MSC therapy group

Fang et al. [55]

I/IIа

18/6/6 EPCs

Autologous MSCs (bone marrow)/

2.5×106 cells + 2.5×106 cells/kg after 1 week

Intravenous transplantation

4 years

No statistically significant differences

between groups

Hess et al. [56]

II

129/67

Allogeneic MSCs (bone marrow)/

400 mln

cells/kg

or 1200 mln cells/kg

Intravenous transplantation

3 years

Positive therapeutic effect observed with early MSC transplantation within 12–36 hours

Levy et al. [57]

I/II

36

Allogeneic MSCs (bone marrow)/

1.5 million

cells/kg

Intravenous transplantation

12 months

Significant increase in mBI after

6 months

and 12 months

Steinberg et al. [58]

I/IIa

18

MSCs (SB623 bone marrow line)/ 2.5×106, 5.0×106 or 10×106 cells

Intrathecal stereotactic

2 years

Meaningful

functional improvement (ESS, NIHSS, F-M) from month 1

Qiao et al. [59]

I

8

Allogeneic MSCs

(umbilical cord)/

0.5×106 cells/kg intravenously

or 5×106 cells

of MSCs + 6×106 cells

of NSCs/NPCs intrathecally

Intrathecal MSC transplantation by 4 injections or co-transplantation of 1 dose of MSC intravenously and MSC + NSC intrathecally

2 years

Functional improvement (NIHSS, mBI, and mRS) more pronounced after

co-transplantation

Jaillard

et al. [60]

I/II

44/31

Autologous MSCs (bone marrow)/

1×106 cells/kg

Intravenous transplantation

90 days

Significant improvement

in F-M scale and restoration

of interhemispheric and ipsilateral conduction pathways

Jaillard

и др. [61]

II

31/16

Autologous MSCs (bone marrow)/

1×108 cells

or 3×108 cells

Intravenous transplantation

2 years

Significant improvement

in motor function

De Celis-

Ruiz et al. [62]

IIb

30/15

Allogeneic MSCs (adipose tissue)/

106 cells/kg

Intravenous transplantation

2 years

Improvement

trend in NIHSS

score

Bang et al. [63]

II

54/39

Autologous MSCs (bone marrow)/

1×106 cells/kg

Intravenous transplantation

90 days

Significant increase (~5-fold) in blood levels of extracellular vesicles and microRNAs associated with neurogenesis/neuroplasticity 24 hours after transplantation, correlating with recovery of motor function and pathway

Note: MSCs — mesenchymal stem cells; EPCs — endothelial progenitor cells; SC — stem cells; NIHSS — National Institutes of Health Stroke Scale; mBI — modified Barthel Index; mRS — modified Rankin scale; ESS — European Stroke Scale; F-M — Fugl-Meyer scale; NSCs/NPCs — neural stem/progenitor cells.

 

Transplantation of autologous MSCs

In 2005, Bang et al. have conducted one of the first CTs on cell therapy for IS using MSC transplantation. The study involved intravenous transplantation of autologous bone marrow MSCs at a dose of 100 million cells (using cell culture media containing fetal bovine serum) in five individuals within 5–7 weeks from the onset of the disease. No serious side effects were found, and the transplantation was deemed safe. Furthermore, patients who received cell therapy demonstrated persistent clinical improvement and a reduction in neurological deficit severity at 3, 6, and 12 months post-transplantation compared with 25 controls.

Following the success of their initial study, Bang et al. have performed a larger blinded phase II study with 52 patients [51]. The study involved intravenous MSC transplantation in two stages: an initial dose of 50 million cells followed by an additional 50 million cells 2 weeks later. Furthermore, the study confirmed the safety of intravenous MSC transplantation and reported that using bovine serum to culture MSCs did not result in zoonosis or any other adverse effects. The cell therapy group showed higher functional recovery and lower mortality than the control group.

Owing to concerns within the scientific community regarding prion disease transmission through animal-derived media and sera used for culturing MSCs, Honmou et al. have conducted a study in which MSCs were cultured in autologous serum [52]. They performed intravenous transplantation of bone marrow-derived MSCs in patients 36–133 days after IS. The study confirmed the safety and feasibility of culturing autologous bone marrow MSCs using autologous human serum. No significant adverse events were detected. In the patients who received cell therapy, a decrease in neurological deficits and significant decrease in the size of the brain infarction focus were observed 1 week after intravenous transplantation.

In 2011, Bhasin et al. transplanted autologous bone marrow MSCs into patients with IS for the first time. MSCs were cultured on serum-free medium. In the phase I/II study, MSCs were administered intravenously to 12 patients with IS in the early and late recovery period (from 3 months to 1 year from disease onset). The data were compared with those of the control group (6 patients). In the group that received MSC cell therapy, the severity of neurological deficits decreased; however, these changes were not statistically significant when compared with those in the control group. In 2013, Bhasin et al. initiated another clinical trial [54] to compare the efficacy of MSCs with that of hematopoietic/mononuclear cells. The results confirmed the safety and feasibility of cell therapy using MSCs. No significant differences were found between the controls and patients who showed improved neurological deficit.

In 2019, Fang et al. [55] showed results of a clinical randomized placebo-controlled phase I/IIa study. The study involved the intravenous transplantation of autologous bone marrow MSCs to patients with IS. MSCs were cultured using fetal bovine serum. The efficacy of the treatment was evaluated 1 week after the onset of the disease. First, bone marrow was collected to isolate and culture autologous MSCs. These cells were then administered intravenously in two stages: the first dose was 2.5 million cells per kilogram of body weight, given after 4 weeks, followed by a second dose of the same amount after 1 week. The study compared therapeutic efficacy and safety between two groups: one group (6 people) received MSC transplantation and the other group (6 people) received a placebo that was intravenously injected with autologous endothelial progenitor cells. The follow-up period was 4 years, and the study was considered safe. No significant differences were found between the two groups regarding survival and degree of functional recovery. One possible reason for the insufficient therapeutic effect may be the introduction of MSCs during the subacute period of IS. To solve this problem, allogeneic stem cells should be transplanted in patients during the acute phase of IS.

Transplantation of allogeneic native MSCs

MSCs can be easily multiplied in culture and weakly express antigens of the major histocompatibility complex HLA-ABC, making their allogeneic transplantation possible [64, 65, 66]. This type of transplantation has advantages for the treatment of neurological diseases. First, it significantly reduces costs and simplifies production, allowing for the creation of a standardized bank of MSCs that can be prepared in advance and used in the acute period of IS as needed. Second, the age of the donors is a crucial factor to consider. Epidemiological studies have indicated that over 75% of all cases of IS occur in individuals aged >65 years [67]. Obtaining MSCs from bone marrow in this age group is challenging, and the regenerative potential of such MSCs is significantly lower than that of MSCs obtained from young donors. This difference is due to natural aging processes [68].

The largest randomized double-blind placebo-controlled phase II MASTERS trial to date, which studied the effect of allogeneic MSC transplantation in IS, was conducted across 33 medical centers in the USA and the UK [56]. This study investigated the safety and efficacy of the MultiStem cell product, which consists of allogeneic bone marrow MSCs obtained from adult donors [69]. Patients were administered MSCs intravenously at a dose of either 400 million or 1.2 billion cells 24–48 h after disease onset. The safety of this technology was confirmed when both doses of MSCs were administered. However, the primary endpoint of achieving the expected degree of improvement in the functional status of patients 90 days after IS was not met when comparing the cell therapy group with the placebo group. A retrospective analysis of the results obtained in some patients with functional recovery still showed statistically significant improvement. The researchers used this data to initiate the next phase of the clinical trial, which is a prospective randomized placebo-controlled double-blind phase III study (MASTERS-2). The study began between 18 and 36 h after the onset of neurological deficit and is currently ongoing. The results have not yet been published.

In a phase I/II study, Levy et al. [57] have evaluated the safety and efficacy of allogeneic bone marrow MSCs obtained from a single healthy donor. MSCs were transplanted intravenously at a dose of up to 1.5 million cells per kilogram of body weight to 38 patients in the late recovery period of IS (>6 months from disease onset). The study revealed the safety of MSC infusion, and all patients demonstrated significant functional recovery according to the Barthel scale. However, this study did not include a control group.

Transplantation of modified allogeneic MSCs

In phase I/IIa, Dr. Steinberg and a team of scientists studied the safety and efficacy of transplanting a modified line of allogeneic bone marrow MSCs, known as SB623, in patients with IS. SB623 is a modified line of bone marrow-derived MSCs. These cells were transiently transfected with a plasmid containing the intracellular domain of Notch1, resulting in Notch1 overexpression. Notch1 plasmid does not replicate during mitosis and is rapidly lost during cell division. In PCSs, Notch1-modified MSCs were found to have a neurotrophic effect, improve survival and maintain neuronal viability in cerebral ischemia, improve neoangiogenesis, and have an anti-inflammatory effect [70–73]. In experimental IS models, SB623 MSCs demonstrated both functional recovery and neuroprotective effects on neurons in the peri-infarct area. However, the transplanted cells did not have a direct substitutive effect and were not preserved in the recipient’s body for an extended period [74]. In CTs, the SB623 cell line was stereotactically injected into the brains of 18 patients during the late recovery period of IS (6 months to 3 years from disease onset) at doses of 2.5, 5, or 10 million cells. Each patient received 20 stereotactic injections in the brain infarction areas. The authors attributed adverse events, such as headache, nausea, and vomiting, to neurosurgery rather than the effects of the transplanted cells. No graft rejection reactions were observed. The study demonstrated significant functional recovery in patients at 3, 6, and 12 months, with sustained clinical improvement 2 years after transplantation [75]. A larger randomized phase IIb clinical trial has been initiated because of the promising results; however, its findings have not yet been published.

Combined transplantation of MSCs and other stem/progenitor cells

In the context of PCSs, MSCs secrete various paracrine factors and exhibit trophic, neuroprotective, anti-inflammatory, and immunomodulatory effects [46, 76]. It is hypothesized that joint transplantation of MSCs with other types of stem/progenitor cells can improve graft engraftment and enhance the therapeutic effects of cell therapy. Animal studies using models of experimental brain infarction have confirmed that combined transplantation of MSCs with neural [77] or endothelial progenitor cells [78] can have a more pronounced effect than monotherapy with only one type of stem/progenitor cells [76]. This may be due to the synergy between the therapeutic effects of the two different stem cell types. Several CTs have been conducted on the combined transplantation of MSCs and other types of stem cells on patients with IS based on encouraging preclinical data.

In a pilot clinical study, Chen et al. [79] transplanted allogeneic MSCs obtained from umbilical cord blood, along with various fetal cells of neural origin (neural progenitor cells, cells of olfactory lining, Schwann cells isolated from sciatic nerve), in 10 patients with ischemic or hemorrhagic stroke in the late recovery period (from 6 months to 20 years). MSCs were transplanted intravenously, whereas the other cell types were injected intracerebrally or intrathecally. The study results demonstrated an improvement in the clinical condition of patients without significant adverse reactions. However, the sample size in each group was negligible.

Qiao et al. [59] have assessed the safety and feasibility of transplanting human fetal neural progenitor cells and allogeneic MSCs isolated from umbilical cord blood in 8 patients with IS at different stages of the disease (ranging from 1 week to 2 years after onset). The first group of patients received four intravenous injections of MSCs at 500,000 cells per kilogram of body weight. The second group received a single intravenous infusion followed by three consecutive injections of MSCs and neural progenitor cells into the brain cisterns. The combined transplantation of MSCs and NPCs was deemed feasible and safe. Each treated patient showed clinical improvement, which was sustained for 2 years after transplantation. No oncogenic transformation of organs was observed during this period.

To confirm these findings, further randomized placebo-controlled multicenter CTs on a larger sample of patients with IS are recommended.

Mechanisms of MSC action

Currently, numerous preclinical studies have investigated the efficacy and mechanisms of MSCs in experimental IS [80]. The following presents generalized data on the therapeutic mechanism of MSCs from an evidence-based medicine perspective.

Although MSCs are multipotent stem cells capable of differentiating into various mesodermal cells, some scientists revealed that MSCs have low or no potential for transdifferentiation in the ectodermal direction, specifically into neural or glial cells in the brain infarction area. Therefore, a direct substitutive mechanism of action is unlikely. Single cases have been described in which, after intracerebral injection, some of the transplanted MSCs lost expression of specific markers and acquired a neuron-like phenotype [81]. Simultaneously, a significant amount of data indicated that the therapeutic potential of MSCs is linked to their paracrine action. This refers to their ability to secrete anti-inflammatory cytokines, growth factors, and exosomes loaded with biologically active substances such as microRNAs, cytokines, and growth factors [82]. Furthermore, several studies have shown that MSCs can fuse with other cells, which may be a mechanism of tissue regeneration [83]. Transplanted MSCs have a direct neuroprotective effect by transferring mitochondrial and cytoplasmic components to the recipient nerve and glial cells [84]. Several studies have reported enhancement of endogenous neurogenesis in recipient animals after MSC transplantation [85–87].

Currently, the therapeutic effect of MSCs is primarily due to their paracrine effect, rather than their ability to differentiate into other cell types [88, 89]. MSCs secrete factors that influence tissue regeneration, reduce inflammation, and promote angiogenesis. These factors include cytokines that modulate the immune response, such as interleukin (IL)-6, and regulatory molecules, including vascular endothelial growth factor (VEGF), IGF-1, IGF-2, and PDGF-AA. These molecules were detected in the conditioned medium in which MSCs were cultured. Moreover, a transcriptome analysis showed that human MSCs differ from fibroblasts, osteoblasts, chondrocytes, and adipocytes, among other types of differentiated cells, because of increased expression of certain molecules. Specifically, the levels of brain-derived neurotrophic factor (BDNF) are approximately 20-fold higher, IL-6 is 60-fold higher, and VEGF is 20-fold higher [90]. Experimental evidence confirms the neuroprotective role of BDNF secreted by human MSCs [91]. Furthermore, MSC transplantation can stimulate recipient cells to produce VEGF and basic fibroblast growth factor, as demonstrated in a rat model of brain ischemia [92]. IL-6 plays a pro-inflammatory role [93] and a crucial role in regeneration and stemness. It helps maintain the “stem” phenotype of MSCs [94] and can activate axon regeneration in mature retinal ganglion cells under certain conditions [95].

Currently, researchers are attempting to determine the molecular composition of biologically active substances in exosomes secreted by MSCs. The exosomes of MSCs from adipose tissue contain 591 proteins, which are mainly involved in signal transduction (GO:0007165), cell adhesion (GO:0007155), positive regulation of proliferation (GO:0008284), and immune response (GO:0006955), according to Gene Ontology [96]. Exosomes contain 489 microRNAs from various families, including the mir-515 and mir-10 families. A significant portion of microRNAs in MSC-secreted exosomes are believed to exist as microRNA precursors [79]. Bioinformatics indicates that some of these precursors are involved in regulating inflammatory processes, such as microRNAs hsa-let-7g-5p, hsa-miR-16-5p, and hsa-miR-92a-3p, or in negatively regulating macrophage activation, such as hsa-miR-124-3p [97]. Furthermore, microRNA targets of secreted MSCs in exosomes include the Wnt signaling pathway, profibrotic signaling cascades such as TGF-β (transforming growth factor-β) and PDGF (platelet-derived growth factor), and signaling pathways that regulate cell proliferation and apoptosis [98].

The immunomodulatory effect of MSCs is based on their ability to influence the polarization of macrophages in the area of injury. This polarization can lead to the M2 phenotype, which is anti-inflammatory, in response to cytokines such as IL-4, IL-10, and IL-13. Polarization plays a crucial role in central nervous system damage. Macrophages, depending on their phenotype, can either contribute to secondary tissue damage or aid in its repair [99]. This process is influenced by the activation of pro-inflammatory M1 or anti-inflammatory M2 macrophages. Research has demonstrated that MSCs can influence the polarization of macrophages toward M2 through regulatory molecule secretion [100]. Remarkably, this polarization is regulated by both cytokines and nucleic acids found in exosomes secreted by MSCs. For instance, a long noncoding RNA called LncGm37494 possesses such a function. It can promote macrophage polarization in the M2 direction by inhibiting miR-130b-3p microRNA and stimulating PPARγ expression [101]. MSCs can influence the balance of T-cell subpopulations, particularly Treg and Th17, as demonstrated in vitro [102]. This effect can lead to the repair of damaged tissue. MSCs also affect Breg cells although the mechanism of this effect is poorly understood [103]. Both Treg and Breg cells are key suppressors of inflammation and autoimmune reactions.

MSCs can affect the state of the extracellular matrix, which is a crucial component of the cellular microenvironment that regulates cell differentiation, migration, and tissue repair. MSCs produce and deposit fibronectin, a component of the extracellular matrix, which can promote tissue regeneration, as demonstrated in a model of spinal cord injury [104]. Additionally, MSCs secrete proangiogenic factors, which have been extensively covered in recent publications. Factors secreted by MSCs include growth factors, such as EGF, FGF-2, ANGPT1, ANG, PDGF, TGF-α, TGF-β, and VEGF [105], and regulatory nucleic acids that affect angiogenesis. Among these nucleic acids are proangiogenic microRNAs, including miR-30b [106].

Strategies for developing cell therapy using MSCs

None of the CTs conducted to date in patients with IS after MSC transplantation have revealed serious adverse events. All studies have noted a tendency toward improvement in patients’ condition and a reduction in the severity of neurological deficits. In studies involving humans, where patients were randomized and a control group was included, not all patients demonstrated a statistically significant positive effect of MSC cell therapy compared with the controls. Possible reasons for insufficiently expressed clinical effects include suboptimal parameters of cell transplantation and criteria for patient inclusion in trials, such as a limited therapeutic window and variation of several months regarding MSC infusion. Moreover, therapy is often performed in the late recovery period of IS, and the comparison group may include patients with a large age difference (from childhood/youth to old age) or those who receive autologous MSCs from elderly donors. The choice of transplantation method and the frequency of MSC administration should be considered. In most studies, MSCs were administered intravenously once or twice during the therapy period. Recent preclinical studies have demonstrated the high efficiency of the intra-arterial method of stem cell administration. This method allows for targeted delivery of transplanted cells to cerebral vessels, bypassing parenchymal organs [44]. Selecting the most effective therapeutic window and transplantation method is dependent on the presumed mechanisms of stem cell action, which require further study.

To further develop cell therapy for IS, one strategy is to modify and unify protocols for MSC use for further optimization. Currently, randomized placebo-controlled multicenter CTs with modified protocols of cell transplantation are underway in several countries. The results of these trials will be available within the next few years [107].

In addition to continuing CTs, an essential strategy for developing cell therapy using MSCs is to continue translational basic research on experimental brain infarction models in laboratory animals. This study aimed to clarify the mechanisms of action of transplanted stem cells and optimize transplantation protocols for their subsequent introduction into clinical practice. When conducting translational studies, evaluating the efficacy of MSC therapy in animals of different genders and ages and in those with concomitant pathologies such as diabetes mellitus and arterial hypertension is advisable [108, 109]. Additionally, investigating the mechanisms of MSC therapeutic action and ways to enhance it is critical. For example, combining MSC transplantation with neuronal progenitor cells obtained through modern methods that do not involve the use of human embryo or fetal tissues [34, 110] may be effective.

To assess the efficacy of MSC cell therapy in preclinical studies, objective quantitative methods of therapy evaluation, in particular the degree of neurological deficit severity, such as kinematics of movements of the paralyzed limb and assessment of the volume of the brain infarction focus, should be used. Wider implementation of new methods at the preclinical level will allow selecting the best ways to objectify the results of cell therapy evaluation and use them in the design of CTs. Evaluation of the brain infarction center volume is a critical objective quantitative parameter for assessing the quality of cell therapy. Morphometric analysis of brain infarction can be performed in experimental studies through histological examination [111, 112] and/or magnetic resonance imaging (MRI) [111, 112]. In preclinical studies on experimental models of cerebral ischemia, MRI allows for the assessment of changes in the volume of the brain infarction without the need to remove animals from the experiment for histologic analysis at each time point. When transferred to computer systems, MRI enables objective assessment of the dynamics of the pathological process in the brain before and after treatment throughout the patient’s lifetime [113–115]. For quantitative assessment of MRI data, a promising method is fully automatic parametric image analysis [116] or convolutional neural networks [117]. These methods minimize the subjective contribution of the operator when assessing the data. However, to increase the objectivity of the assessment, the process of morphometric analysis of the brain infarction focus should be standardized for optimal use upon the patient’s hospital admission. One of the methods is the segmentation method [118] by selecting the region of interest on a series of images. This approach allows avoiding errors due to the subjectivity of focal volume assessment by an operator or several operators without averaging their assessment.

The development and testing of methods for automatic objective assessment of cell therapy efficacy in cerebral infarction in PCSs and CTs can create a solid foundation for qualitative assessment of the results obtained.

CONCLUSIONS

Based on the analysis of the conducted CTs of the safety and efficacy of cell therapy for IS, it can be concluded that MSC transplantation is a safe and effective procedure from a pathogenetic perspective.

Continuing research in this direction, including the initiation of the first CTs in Russia, is recommended. To introduce IS therapy into clinical practice, CTs on a large sample of patients with randomization and adequate selection of a control group should be conducted. This should include criteria modification for patient inclusion in the study and protocols of MSC transplantation corresponding to a high degree of evidence. Further fundamental research on the mechanisms of cell therapy action and the selection of the optimal time window, methods, and frequency of stem cell administration is warranted.

ADDITIONAL INFORMATION

Funding source. This work was supported by the Russian Science Foundation (project No. 23-25-00300). Work by D.A. Chudakova carried out with the support of the Federal Medical and Biological Agency of Russia.

Competing interests. The authors declare that they have no competing interests.

Authors’ contribution. D.D. Namestnikova, D.B. Kovalenko, I.A. Pokusaeva, D.A. Chudakova — data collection and analysis, manuscript writing, editing, research concept and design; I.L. Gubskiy, K.N. Yarygin, V.P. Baklaushev — manuscript writing, editing, research concept and design. The authors made a substantial contribution to the conception of the work, acquisition, analysis, interpretation of data for the work, drafting and revising the work, final approval of the version to be published and agree to be accountable for all aspects of the work.

×

About the authors

Daria D. Namestnikova

Federal Center of Brain Research and Neurotechnologies; Pirogov Russian National Research Medical University

Author for correspondence.
Email: dadnam89@gmai.com
ORCID iD: 0000-0001-6635-511X
SPIN-code: 1576-1860

MD, PhD, Research Associate

Russian Federation, Moscow; Moscow

Daria B. Kovalenko

Pirogov Russian National Research Medical University

Email: daarrr.iii01@gmail.com
Russian Federation, Moscow

Irina A. Pokusaeva

Pirogov Russian National Research Medical University

Email: pokusaeva.i@yandex.ru
Russian Federation, Moscow

Daria A. Chudakova

Federal Center of Brain Research and Neurotechnologies

Email: daria.chd@gmail.com
ORCID iD: 0000-0002-9354-6824
SPIN-code: 1410-9581

PhD, Senior Research Associate

Russian Federation, Moscow

Ilya L. Gubskiy

Federal Center of Brain Research and Neurotechnologies; Pirogov Russian National Research Medical University

Email: gubskiy.ilya@gmail.com
ORCID iD: 0000-0003-1726-6801
SPIN-code: 9181-3091

MD, PhD, Senior Research Associate

Russian Federation, Moscow; Moscow

Konstantin N. Yarygin

Institute of Biomedical Chemistry

Email: kyarygin@yandex.ru
ORCID iD: 0000-0002-2261-851X
SPIN-code: 7567-1230

PhD, Professor

Russian Federation, Moscow

Vladimir P. Baklaushev

Federal Center of Brain Research and Neurotechnologies; Pirogov Russian National Research Medical University; Federal Scientific and Clinical Center for Specialized Medical Assistance and Medical Technologies of the Federal Medical Biological Agency

Email: baklaushev.vp@fnkc-fmba.ru
ORCID iD: 0000-0003-1039-4245
SPIN-code: 3968-2971

PhD, Assistant Professor

Russian Federation, Moscow; Moscow; Moscow

References

  1. Kim J, Thayabaranathan T, Donnan GA, et al. Global Stroke Statistics 2019. Int J Stroke. 2020;15(8):819–838. doi: 10.1177/1747493020909545
  2. Pu L, Wang L, Zhang R, et al. Projected global trends in ischemic stroke incidence, deaths and disability-adjusted life years from 2020 to 2030. Stroke. 2023;54(5):1330–1339. doi: 10.1161/STROKEAHA.122.040073
  3. Гудкова В.В., Кимельфельд Е.И., Белов С.Е. Отек головного мозга: от истоков описания к современному пониманию процесса // Consil Med. 2021. Т. 23, № 2. С. 131–135. [Gudkova VV, Kimelfeld EI, Belov SE. Brain oedema: From the origins of description to the modern understanding of the process. Consil Med. 2021;23(2):131–135. (In Russ).] doi: 10.26442/20751753.2021.2.200604
  4. Игнатьева В.И., Вознюк И.А., Шамалов Н.А., и др. Социально-экономическое бремя инсульта в Российской Федерации // Журнал неврологии и психиатрии им. С.С. Корсакова. 2023. Т. 123, № 8. С. 5. [Ignatyeva VI, Voznyuk IA, Shamalov NA, et al. Social and economic burden of stroke in Russian Federation. J Neurol Psychiatry named after S.S. Korsakov. 2023;123(8):5. (In Russ).] doi: 10.17116/jnevro20231230825
  5. Feigin VL, Stark BA, Johnson CO, et al. Global, regional, and national burden of stroke and its risk factors, 1990–2019: A systematic analysis for the Global Burden of Disease Study 2019. Lancet Neurol. 2021;20(10):795–820. doi: 10.1016/S1474-4422(21)00252-0
  6. The top 10 causes of death [Electronic resource]. Режим доступа: https://www.who.int/en/news-room/fact-sheets/detail/the-top-10-causes-of-death. Дата обращения: 09.05.2022.
  7. Число умерших по основным классам причин смерти. Демография. Федеральная служба государственной статистики [Electronic resource]. Режим доступа: https://rosstat.gov.ru/folder/12781#. Дата обращения: 08.11.2023.
  8. Солдатов М.А., Климов Л.В., Толмачев А.П., и др. Внутривенная тромболитическая терапия ишемического инсульта препаратом Ревелиза в реальной клинической практике: Результаты исследования IVT-AIS-R // Журнал неврологии и психиатрии им. С.С. Корсакова. 2022. Т. 122, № 12. С. 42. [Soldatov MA, Klimov LV, Tolmachev AP, et al. Intravenous thrombolytic therapy of ischemic stroke with the drug Revelisa in real clinical practice: Results of the IVT-AIS-R study. J Neurol Psychiatry named after S.S. Korsakov. 2022;122(12):42. (In Russ).] doi: 10.17116/jnevro202212212242
  9. Акжигитов Р.Г., Алекян Б.Г., Алферова В.В. Ишемический инсульт и транзиторная ишемическая атака у взрослых. Клинические рекомендации. Москва, 2021. 181 c. [Akzhigitov RG, Alekian BG, Alferova VV. Ischaemic stroke and transient ischaemic attack in adults. Clinical Recommendations. Moscow; 2021. 181 р. (In Russ).]
  10. Powers WJ, Rabinstein AA, Ackerson T, et al. Guidelines for the early management of patients with acute ischemic stroke: 2019 update to the 2018 guidelines for the early management of acute ischemic stroke a guideline for healthcare professionals from the American Heart Association/American Stroke. Stroke. 2019;50(12):E344–E418. doi: 10.1161/STR.0000000000000211
  11. Nogueira RG, Jadhav AP, Haussen DC, et al. Thrombectomy 6 to 24 hours after stroke with a mismatch between deficit and infarct. New Engl J Med. 2018;378(1):11–21. doi: 10.1056/NEJMoa1706442
  12. Федин А.И., Бадалян К.Р. Обзор клинических рекомендаций по лечению и профилактике ишемического инсульта // Журнал неврологии и психиатрии им. С.С. Корсакова. 2019. Т. 119, № 8. C. 95–100. [Fedin AI, Badalyan KR. Review of clinical guidelines for the treatment and prevention of ischemic stroke. J Neurol Psychiatry named after S.S. Korsakov. 2019;119(8): 95–100. (In Russ).] doi: 10.17116/jnevro201911908295
  13. Гусев Е.И., Мартынов М.Ю., Ясаманова А.Н. Тромболитическая терапия ишемического инсульта // Журнал неврологии и психиатрии им. C.C. Корсакова. 2018. Т. 118, № 12-2. C. 4–14. [Gusev EI, Martynov MY, Yasamanova AN. Thrombolytic therapy of ischaemic stroke. J Neurol Psychiatry named after S.S. Korsakov. 2018;118 (12-2):4–14. (In Russ).] doi: 10.17116/jnevro20181181224
  14. Horwitz EM, Le Blanc K, Dominici M, et al. Clarification of the nomenclature for MSC: The international society for cellular therapy position statement. Cytotherapy. 2005;7(5):393–395. doi: 10.1080/14653240500319234
  15. Jin QH, Kim HK, Na JY, et al. Anti-inflammatory effects of mesenchymal stem cell-conditioned media inhibited macrophages activation in vitro. Sci Rep. 2022;12(1):4754. doi: 10.1038/s41598-022-08398-4
  16. Pang QM, Chen SY, Fu SP, et al. Regulatory role of mesenchymal stem cells on secondary inflammation in spinal cord injury. J Inflammat Res. 2022;(15):573–593. doi: 10.2147/JIR.S349572
  17. Dominici M, Le Blanc K, Mueller I, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The international society for cellular therapy position statement. Cytotherapy. 2006;8(4):315–317. doi: 10.1080/14653240600855905
  18. Mikael PE, Willard C, Koyee A, et al. Remodeling of glycosaminoglycans during differentiation of adult human bone mesenchymal stromal cells toward hepatocytes. Stem Cells Development. 2019;28(4):278–289. doi: 10.1089/scd.2018.0197
  19. Gao Q, Guo M, Jiang X, et al. A Cocktail method for promoting cardiomyocyte differentiation from bone marrow-derived mesenchymal stem cells. Stem Cells Int. 2014;2014:1–11. doi: 10.1155/2014/162024
  20. Scuteri A, Miloso M, Foudah D, et al. Mesenchymal stem cells neuronal differentiation ability: A real perspective for nervous system repair? Curr Stem Cell Res Therapy. 2011;6(2):82–92. doi: 10.2174/157488811795495486
  21. Lee BB, Cripps RA, Fitzharris M, et al. The global map for traumatic spinal cord injury epidemiology: Update 2011, global incidence rate. Spinal Cord. 2014;52(2):110–116. doi: 10.1038/sc.2012.158
  22. Gartner S, Kaplan HS. Long-term culture of human bone marrow cells. Proc Natl Acad Sci USA. 1980;77(8):4756–4759. doi: 10.1073/pnas.77.8.4756
  23. Kim DW, Staples M, Shinozuka K, et al. Wharton’s jelly-derived mesenchymal stem cells: Phenotypic characterization and optimizing their therapeutic potential for clinical applications. Int J Mol Sci. 2013;14(6):11692–11712. doi: 10.3390/ijms140611692
  24. Fei X, Jiang S, Zhang S, et al. Isolation, culture, and identification of amniotic fluid-derived mesenchymal stem cells. Cell Biochem Biophys. 2013;67(2):689–694. doi: 10.1007/s12013-013-9558-z
  25. Secunda R, Vennila R, Mohanashankar AM, et al. Isolation, expansion and characterisation of mesenchymal stem cells from human bone marrow, adipose tissue, umbilical cord blood and matrix: A comparative study. Cytotechnology. 2015;67(5): 793–807. doi: 10.1007/s10616-014-9718-z
  26. Pierdomenico L, Bonsi L, Calvitti M, et al. Multipotent mesenchymal stem cells with immunosuppressive activity can be easily isolated from dental pulp. Transplantation. 2005;80(6):836–842. doi: 10.1097/01.tp.0000173794.72151.88
  27. Elahi KC, Klein G, Avci-Adali M, et al. Human mesenchymal stromal cells from different sources diverge in their expression of cell surface proteins and display distinct differentiation patterns. Stem Cells Int. 2016;2016:5646384. doi: 10.1155/2016/5646384
  28. Griffin MD, Ritter T, Mahon BP. Immunological aspects of allogeneic mesenchymal stem cell therapies. Hum Gene Therapy. 2010;21(12):1641–1655. doi: 10.1089/hum.2010.156
  29. Kenmuir CL, Wechsler LR. Update on cell therapy for stroke. Stroke Vasc Neurol. 2017;2(2):59–64. doi: 10.1136/svn-2017-000070
  30. Neri S. Genetic stability of mesenchymal stromal cells for regenerative medicine applications: A fundamental biosafety aspect. Int J Mol Sci. 2019;20(10):2406. doi: 10.3390/ijms20102406
  31. Наместникова Д.Д., Таирова Р.Т., Сухинич К.К., и др. Клеточная терапия ишемического инсульта. Типы стволовых клеток и результаты доклинических испытаний // Журнал неврологии и психиатрии имени С.С. Корсакова. 2018. Т. 118, № 9. С. 69–75. [Namestnikova DD, Tairova RT, Sukhinich KK, et al. Cell therapy for ischemic stroke. Stem cell types and results of preclinical trials. J Neurol Psychiatry named after S.S. Korsakov. 2018;118(9):69–75. (In Russ).] doi: 10.17116/jnevro201811809269
  32. Rascón-Ramírez FJ, Esteban-García N, Barcia JA, et al. Are we ready for cell therapy to treat stroke? Fron Cell Dev Biol. 2021;(9):621645. doi: 10.3389/fcell.2021.621645
  33. Cherkashova E, Namestnikova D, Leonov G, et al. Comparative study of the efficacy of intra-arterial and intravenous transplantation of human induced pluripotent stem cells-derived neural progenitor cells in experimental stroke. Peer J. 2023;(11):e16358. doi: 10.7717/peerj.16358
  34. Namestnikova DD, Gubskiy IL, Revkova VA, et al. Intra-arterial stem cell transplantation in experimental stroke in rats: Real-time MR visualization of transplanted cells starting with their first pass through the brain with regard to the therapeutic action. Front Neurosci. 2021;15:641970. doi: 10.3389/fnins.2021.641970
  35. Sukhinich KK, Namestnikova DD, Gubskii IL, et al. Distribution and migration of human placental mesenchymal stromal cells in the brain of healthy rats after stereotaxic or intra-arterial transplantation. Bulletin Exp Biol Med. 2020;168(4):542–551. doi: 10.1007/s10517-020-04750-8
  36. Cherkashova EA, Burunova VV, Bukharova TB, et al. Comparative analysis of the effects of intravenous administration of placental mesenchymal stromal cells and neural progenitor cells derived from induced pluripotent cells on the course of acute ischemic stroke in rats. Bulletin Exp Biol Med. 2019;166(4):558–566. doi: 10.1007/s10517-019-04392-5
  37. Cherkashova EA, Namestnikova DD, Gubskiy IL, et al. Dose-dependent effects of intravenous mesenchymal stem cell transplantation in rats with acute focal cerebral ischemia. Bulletin Exp Biol Med. 2022;173(4):514–518. doi: 10.1007/S10517-022-05573-5
  38. Cherkashova EA, Namestnikova DD, Gubskiy IL, et al. Dynamic MRI of the mesenchymal stem cells distribution during intravenous transplantation in a rat model of ischemic stroke. Life. 2023;13(2):288. doi: 10.3390/life13020288
  39. Namestnikova DD, Gubskiy IL, Cherkashova EA, et al. Therapeutic efficacy and migration of mesenchymal stem cells after intracerebral transplantation in rats with experimental ischemic stroke. Bulletin Exp Biol Med. 2023;175(1):116–125. doi: 10.1007/s10517-023-05822-1
  40. Zhang XL, Zhang XG, Huang YR, et al. Stem cell-based therapy for experimental ischemic stroke: A preclinical systematic review. Front Cell Neurosci. 2021;(15):628908. doi: 10.3389/fncel.2021.628908
  41. Barbash IM, Chouraqui P, Baron J, et al. Systemic delivery of bone marrow-derived mesenchymal stem cells to the infarcted myocardium: Feasibility, cell migration, and body distribution. Circulation. 2003;108(7):863–868. doi: 10.1161/01.CIR.0000084828.50310.6A
  42. Boltze J, Arnold A, Walczak P, et al. The dark side of the force: Constraints and complications of cell therapies for stroke. Front Neurol. 2015;(6):155. doi: 10.3389/fneur.2015.00155
  43. Vasconcelos-dos-Santos A, Rosado-de-Castro PH, Lopes de Souza SA, et al. Intravenous and intra-arterial administration of bone marrow mononuclear cells after focal cerebral ischemia: Is there a difference in biodistribution and efficacy? Stem Cell Res. 2012;9(1):1–8. doi: 10.1016/j.scr.2012.02.002
  44. Guzman R, Janowski M, Walczak P. Intra-arterial delivery of cell therapies for stroke. Stroke. 2018;49(5):1075–1082. doi: 10.1161/STROKEAHA.117.018288
  45. Toyoshima A, Yasuhara T, Kameda M, et al. Intra-Arterial transplantation of allogeneic mesenchymal stem cells mounts neuroprotective effects in a transient ischemic stroke model in rats: Analyses of therapeutic time window and its mechanisms. PLoS One. 2015;10(6):e0127302. doi: 10.1371/journal.pone.0127302
  46. Li W, Shi L, Hu B, et al. Mesenchymal stem cell-based therapy for stroke: Current understanding and challenges. Front Cell Neurosci. 2021;(15):628940. doi: 10.3389/fncel.2021.628940
  47. Zhou L, Zhu H, Bai X, et al. Potential mechanisms and therapeutic targets of mesenchymal stem cell transplantation for ischemic stroke. Stem Cell Res Ther. 2022;13(1):195. doi: 10.1186/S13287-022-02876-2
  48. Zhuang WZ, Lin YH, Su LJ, et al. Mesenchymal stem/stromal cell-based therapy: Mechanism, systemic safety and biodistribution for precision clinical applications. J Biomed Sci. 2021;28(1):28. doi: 10.1186/s12929-021-00725-7
  49. Yong KW, Choi JR, Mohammadi M, et al. Mesenchymal stem cell therapy for ischemic tissues. Stem Cells Int. 2018;2018:8179075. doi: 10.1155/2018/8179075
  50. Bang OY, Lee JS, Lee PH, et al. Autologous mesenchymal stem cell transplantation in stroke patients. Ann Neurol. 2005;57(6):874–882. doi: 10.1002/ana.20501
  51. Lee JS, Hong JM, Moon GJ, et al. A long-term follow-up study of intravenous autologous mesenchymal stem cell transplantation in patients with ischemic stroke. Stem Cells. 2010;28(6): 1099–1106. doi: 10.1002/stem.430
  52. Honmou O, Houkin K, Matsunaga T, et al. Intravenous administration of auto serum-expanded autologous mesenchymal stem cells in stroke. Brain. 2011;134(6): 1790–1807. doi: 10.1093/brain/awr063
  53. Bhasin A, Srivastava MV, Kumaran SS, et al. Autologous mesenchymal stem cells in chronic stroke. Cerebrovasc Dis Extra. 2011;1(1):93–104. doi: 10.1159/000333381
  54. Bhasin A, Srivastava MV, Mohanty S, et al. Stem cell therapy: A clinical trial of stroke. Clin Neurol Neurosur. 2013;115(7): 1003–1008. doi: 10.1016/j.clineuro.2012.10.015
  55. Fang J, Guo Y, Tan S, et al. Autologous endothelial progenitor cells transplantation for acute ischemic stroke: A 4-Year follow-up study. Stem Cells Translat Med. 2019;8(1):14–21. doi: 10.1002/sctm.18-0012
  56. Hess DC, Wechsler LR, Clark M, et al. Safety and efficacy of multipotent adult progenitor cells in acute ischaemic stroke (MASTERS): A randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Neurol. 2017;16(5):360–368. doi: 10.1016/S1474-4422(17)30046-7
  57. Levy ML, Crawford JR, Dib N, et al. Phase I/II study of safety and preliminary efficacy of intravenous allogeneic mesenchymal stem cells in chronic stroke. Stroke. 2019;50(10):2835–2841. doi: 10.1161/STROKEAHA.119.026318
  58. Steinberg GK, Kondziolka D, Wechsler LR, et al. Clinical outcomes of transplanted modified bone marrow-derived mesenchymal stem cells in stroke: A phase 1/2a study. Stroke. 2016;47(7):1817–1824. doi: 10.1161/STROKEAHA.116.012995
  59. Qiao LY, Huang FJ, Zhao M, et al. A two-year follow-up study of cotransplantation with neural stem/progenitor cells and mesenchymal stromal cells in ischemic stroke patients. Cell Transplant. 2014;23(1 Suppl):65–72. doi: 10.3727/096368914x684961
  60. Lee J, Chang WH, Chung W, et al. Efficacy of intravenous mesenchymal stem cells for motor recovery after ischemic stroke: A neuroimaging study. Stroke. 2022;53(1):20–28. doi: 10.1161/STROKEAHA.121.034505
  61. Jaillard A, Hommel M, Moisan A, et al. Autologous mesenchymal stem cells improve motor recovery in subacute ischemic stroke: A randomized clinical trial. Translat Stroke Res. 2020;11(5): 910–923. doi: 10.1007/s12975-020-00787-z
  62. De Celis-Ruiz E, Fuentes B, Moniche F, et al. Allogeneic adipose tissue-derived mesenchymal stem cells in ischaemic stroke (AMASCIS-02): A phase IIb, multicentre, double-blind, placebo-controlled clinical trial protocol. BMJ Open. 2021;11(8):e051790. doi: 10.1136/bmjopen-2021-051790
  63. Bang OY, Kim EH, Cho YH, et al. Circulating extracellular vesicles in stroke patients treated with mesenchymal stem cells: A biomarker analysis of a randomized trial. Stroke. 2022;53(7):2276–2286. doi: 10.1161/STROKEAHA.121.036545
  64. Wang Y, Huang J, Gong L, et al. The plasticity of mesenchymal stem cells in regulating surface HLA-I. iScience. 2019;(15): 66–78. doi: 10.1016/j.isci.2019.04.011
  65. Lee HJ, Kang KS, Kang SY, et al. Immunologic properties of differentiated and undifferentiated mesenchymal stem cells derived from umbilical cord blood. J Veterinary Sci. 2016;17(3):289–297. doi: 10.4142/jvs.2016.17.3.289
  66. Le Blanc K, Tammik C, Rosendahl K, et al. HLA expression and immunologic properties of differentiated and undifferentiated mesenchymal stem cells. Exp Hematol. 2003;31(10):890–896. doi: 10.1016/S0301-472X(03)00110-3
  67. Yousufuddin M, Young N. Aging and ischemic stroke. Aging. 2019;11(9):2542–2544. doi: 10.18632/aging.101931
  68. Yamaguchi S, Horie N, Satoh K, et al. Age of donor of human mesenchymal stem cells affects structural and functional recovery after cell therapy following ischaemic stroke. J Cerebral Blood Flow Metabol. 2018;38(7):1199–1212. doi: 10.1177/0271678X17731964
  69. Boozer S, Lehman N, Lakshmipathy U, et al. Global characterization and genomic stability of human multistem, a multipotent adult progenitor cell. Stem Cell Res Adv. 2011; 4(1):119–134. PMID: 20498688
  70. Dao MA, Tate CC, Aizman I, et al. Comparing the immunosuppressive potency of naïve marrow stromal cells and Notch-transfected marrow stromal cells. J Neuroinflammat. 2011;8(1):133. doi: 10.1186/1742-2094-8-133
  71. Dao M, Tate CC, McGrogan M, et al. Comparing the angiogenic potency of naïve marrow stromal cells and Notch-transfected marrow stromal cells. J Translat Med. 2013;11(1):81. doi: 10.1186/1479-5876-11-81
  72. Tate CC, Fonck C, McGrogan M, et al. Human mesenchymal stromal cells and their derivative, SB623 cells, rescue neural cells via trophic support following in vitro ischemia. Cell Transplant. 2010;19(8):973–984. doi: 10.3727/096368910X494885
  73. Aizman I, Tate CC, McGrogan M, et al. Extracellular matrix produced by bone marrow stromal cells and by their derivative, SB623 cells, supports neural cell growth. J Neurosci Res. 2009;87(14):3198–3206. doi: 10.1002/jnr.22146
  74. Yasuhara T, Matsukawa N, Hara K, et al. Notch-Induced rat and human bone marrow stromal cell grafts reduce ischemic cell loss and ameliorate behavioral deficits in chronic stroke animals. Stem Cells Development. 2009;18(10):1501–1514. doi: 10.1089/scd.2009.0011
  75. Steinberg GK, Kondziolka D, Wechsler LR, et al. Two-year safety and clinical outcomes in chronic ischemic stroke patients after implantation of modified bone marrow–derived mesenchymal stem cells (SB623): A phase 1/2a study. J Neurosur. 2019; 131(5):1462–1472. doi: 10.3171/2018.5.JNS173147
  76. Namestnikova DD, Cherkashova EA, Sukhinich KK, et al. Combined cell therapy in the treatment of neurological disorders. Biomed. 2020;8(12):613. doi: 10.3390/biomedicines8120613
  77. Hosseini SM, Farahmandnia M, Razi Z, et al. Combination cell therapy with mesenchymal stem cells and neural stem cells for brain stroke in rats. Int J Stem Cells. 2015;8(1):99–105. doi: 10.15283/ijsc.2015.8.1.99
  78. Sun K, Zhou Z, Ju X, et al. Combined transplantation of mesenchymal stem cells and endothelial progenitor cells for tissue engineering: A systematic review and meta-analysis. Stem Cell Res Ther. 2016;7(1):151. doi: 10.1186/s13287-016-0390-4
  79. Chen TS, Lai RC, Lee MM, et al. Mesenchymal stem cell secretes microparticles enriched in pre-microRNAs. Nucleic Acids Res. 2009;38(1):215–224. doi: 10.1093/nar/gkp857
  80. Zhang Y, Dong N, Hong H, et al. Mesenchymal stem cells: Therapeutic mechanisms for stroke. Int J Mol Sci. 2022;23(5):2550. doi: 10.3390/ijms23052550
  81. Ullah M, Liu DD, Thakor AS. Mesenchymal stromal cell homing: Mechanisms and strategies for improvement. iScience. 2019;(15):421–438. doi: 10.1016/j.isci.2019.05.004
  82. Harrell C, Fellabaum C, Jovicic N, et al. Molecular mechanisms responsible for therapeutic potential of mesenchymal stem cell-derived secretome. Cells. 2019;8(5):467. doi: 10.3390/cells8050467
  83. Dörnen J, Dittmar T. The role of MSCs and cell fusion in tissue regeneration. Int J Mol Sci. 2021;22(20):10980. doi: 10.3390/ijms222010980
  84. Babenko VA, Silachev DN, Popkov VA, et al. Miro1 enhances mitochondria transfer from multipotent mesenchymal stem cells (MMSC) to neural cells and improves the efficacy of cell recovery. Molecules. 2018;23(3):687. doi: 10.3390/molecules23030687
  85. Chen J, Li Y, Katakowski M, et al. Intravenous bone marrow stromal cell therapy reduces apoptosis and promotes endogenous cell proliferation after stroke in female rat. J Neurosci Res. 2003;73(6):778–786. doi: 10.1002/jnr.10691
  86. Esneault E, Pacary E, Eddi D, et al. Combined therapeutic strategy using erythropoietin and mesenchymal stem cells potentiates neurogenesis after transient focal cerebral ischemia in rats. J Cerebral Blood Flow Metabol. 2008;28(9):1552–1563. doi: 10.1038/jcbfm.2008.40
  87. Maltman DJ, Hardy SA, Przyborski SA. Role of mesenchymal stem cells in neurogenesis and nervous system repair. Neurochem Int. 2011;59(3):347–356. doi: 10.1016/j.neuint.2011.06.008
  88. Caplan AI, Dennis JE. Mesenchymal stem cells as trophic mediators. J Cell Biochem. 2006;98(5):1076–1084. doi: 10.1002/jcb.20886
  89. Lee RH, Pulin AA, Seo MJ, et al. Intravenous hMSCs improve myocardial infarction in mice because cells embolized in lung are activated to secrete the anti-inflammatory protein TSG-6. Cell Stem Cell. 2009;5(1):54–63. doi: 10.1016/j.stem.2009.05.003
  90. Kubo H, Shimizu M, Taya Y, et al. Identification of mesenchymal stem cell (MSC)‐transcription factors by microarray and knockdown analyses, and signature molecule‐marked MSC in bone marrow by immunohistochemistry. Genes Cells. 2009;14(3):407–424. doi: 10.1111/j.1365-2443.2009.01281.x
  91. Wilkins A, Kemp K, Ginty M, et al. Human bone marrow-derived mesenchymal stem cells secrete brain-derived neurotrophic factor which promotes neuronal survival in vitro. Stem Cell Res. 2009;3(1):63–70. doi: 10.1016/j.scr.2009.02.006
  92. Wakabayashi K, Nagai A, Sheikh AM, et al. Transplantation of human mesenchymal stem cells promotes functional improvement and increased expression of neurotrophic factors in a rat focal cerebral ischemia model. J Neurosci Res. 2009;88(5):1017–1025. doi: 10.1002/jnr.22279
  93. Sterner RC, Sterner RM. Immune response following traumatic spinal cord injury: Pathophysiology and therapies. Front Immunol. 2022;(13):1084101. doi: 10.3389/fimmu.2022.1084101
  94. Pricola KL, Kuhn NZ, Haleem‐Smith H, et al. Interleukin‐6 maintains bone marrow‐derived mesenchymal stem cell stemness by an ERK1/2‐dependent mechanism. J Cell Biochem. 2009;108(3):577–588. doi: 10.1002/jcb.22289
  95. Leibinger M, Müller A, Gobrecht P, et al. Interleukin-6 contributes to CNS axon regeneration upon inflammatory stimulation. Cell Death Dis. 2013;4(4):e609–e609. doi: 10.1038/cddis.2013.126
  96. Gene Ontology Resource [Electronic resource]. Режим доступа: https://geneontology.org/. Дата обращения: 12.11.2023.
  97. Alonso-Alonso ML, García-Posadas L, Diebold Y. Extracellular vesicles from human adipose-derived mesenchymal stem cells: A review of common cargos. Stem Cell Rev Rep. 2022;18(3): 854–901. doi: 10.1007/s12015-021-10155-5
  98. Ferguson SW, Wang J, Lee CJ, et al. The microRNA regulatory landscape of MSC-derived exosomes: A systems view. Sci Rep. 2018;8(1):1419. doi: 10.1038/s41598-018-19581-x
  99. Kong X, Gao J. Macrophage polarization: A key event in the secondary phase of acute spinal cord injury. J Cell Mol Med. 2017;21(5):941–954. doi: 10.1111/jcmm.13034
  100. An N, Yang J, Wang H, et al. Mechanism of mesenchymal stem cells in spinal cord injury repair through macrophage polarization. Cell Biosci. 2021;11(1):41. doi: 10.1186/s13578-021-00554-z
  101. Shao M, Jin M, Xu S, et al. Exosomes from long noncoding RNA-Gm37494-ADSCs repair spinal cord injury via shifting microglial M1/M2 polarization. Inflammat. 2020;43(4): 1536–1547. doi: 10.1007/s10753-020-01230-z
  102. Chen QH, WF, Liu L, et al. Mesenchymal stem cells regulate the Th17/Treg cell balance partly through hepatocyte growth factor in vitro. Stem Cell Res Ther. 2020;11(1):91. doi: 10.1186/s13287-020-01612-y
  103. Liu J, Liu Q, Chen X. The immunomodulatory effects of mesenchymal stem cells on regulatory B cells. Front Immunol. 2020;(11):1843. doi: 10.3389/fimmu.2020.01843
  104. Zeng X, Ma Y, Chen Y, et al. Autocrine fibronectin from differentiating mesenchymal stem cells induces the neurite elongation in vitro and promotes nerve fiber regeneration in transected spinal cord injury. J Biomed Materials Res A. 2016; 104(8):1902–1911. doi: 10.1002/jbm.a.35720
  105. Nazari-Shafti TZ, Neuber S, Garcia Duran A, et al. Human mesenchymal stromal cells and derived extracellular vesicles: Translational strategies to increase their proangiogenic potential for the treatment of cardiovascular disease. Stem Cells Translat Med. 2020;9(12):1558–1569. doi: 10.1002/sctm.19-0432
  106. Gong M, Yu B, Wang J, et al. Mesenchymal stem cells release exosomes that transfer miRNAs to endothelial cells and promote angiogenesis. Oncotarget. 2017;8(28):45200–45212. doi: 10.18632/oncotarget.16778
  107. Colliander R, Alleman K, Diaz M, et al. Stem cell implants: Emerging innovation for stroke recovery. J Neuro Oncol Res. 2023;3(1):3102.
  108. Buga M, Di Napoli M, Popa-Wagner A. Preclinical models of stroke in aged animals with or without comorbidities: Role of neuroinflammation. Biogerontol. 2013;14(6):651–662. doi: 10.1007/s10522-013-9465-0
  109. Sommer CJ. Ischemic stroke: Experimental models and reality. Acta Neuropathol. 2017;133(2):245–261. doi: 10.1007/s00401-017-1667-0
  110. Ahlfors JE, Azimi A, El-Ayoubi R, et al. Examining the fundamental biology of a novel population of directly reprogrammed human neural precursor cells. Stem Cell Res Ther. 2019;10(1):166. doi: 10.1186/s13287-019-1255-4
  111. Popp A, Jaenisch N, Witte OW, et al. Identification of ischemic regions in a rat model of stroke. PLoS One. 2009;4(3):e4764. doi: 10.1371/journal.pone.0004764
  112. Weber RZ, Bernardoni D, Rentsch NH, et al. Visualization and estimation of stroke infarct volumes in rodents. bioRxiv. 2023;2023:547245. doi: 10.1101/2023.07.14.547245
  113. Saunders DE, Clifton AG, Brown MM. Measurement of infarct size using MRI predicts prognosis in middle cerebral artery infarction. Stroke. 1995;26(12):2272–2276. doi: 10.1161/01.STR.26.12.2272
  114. González RG. Clinical MRI of acute ischemic stroke. J Magnetic Resonance Imaging. 2012;36(2):259–271. doi: 10.1002/jmri.23595
  115. Milidonis X, Marshall I, Macleod MR, et al. Magnetic resonance imaging in experimental stroke and comparison with histology: Systematic review and meta-analysis. Stroke. 2015;46(3): 843–851. doi: 10.1161/STROKEAHA.114.007560
  116. Qiao J, Cai X, Xiao Q, et al. Data on MRI brain lesion segmentation using K-means and gaussian mixture model-expectation maximization. Data Brief. 2019;27:104628. doi: 10.1016/j.dib.2019.104628
  117. Valverde JM, Shatillo A, De Feo R, et al. Automatic cerebral hemisphere segmentation in rat MRI with ischemic lesions via attention-based convolutional neural networks. Neuroinformat. 2023;21(1):57–70. doi: 10.1007/s12021-022-09607-1
  118. Kevin Zhou S, Fichtinger G, Rueckert D. Handbook of medical image computing and computer assisted intervention. Elsevier; 2019. 1043 p. doi: 10.1016/C2017-0-04608-6

Supplementary files

Supplementary Files
Action
1. JATS XML

Copyright (c) 2024 Eco-Vector

Creative Commons License
This work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.

СМИ зарегистрировано Федеральной службой по надзору в сфере связи, информационных технологий и массовых коммуникаций (Роскомнадзор).
Регистрационный номер и дата принятия решения о регистрации СМИ: серия ПИ № ФС 77 - 38032 от 11 ноября 2009 года.


This website uses cookies

You consent to our cookies if you continue to use our website.

About Cookies