search for




 

Human Endometrial Regenerative Cells for Neurological Disorders: Hype or Hope?
International Journal of Stem Cells 2024;17:224-235
Published online August 30, 2024;  
© 2024 Korean Society for Stem Cell Research.

Javad Momeni1,2,3,4,*, Elnaz Naserzadeh4,*, Ali Sepehrinezhad5, Rezan Ashayeri Ahmadabad3, Sajad Sahab Negah1,2,3

1Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
2Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
3Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
4Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
5Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
Correspondence to: Sajad Sahab Negah
Neuroscience Research Center, Mashhad University of Medical Sciences, Pardis Campus, Azadi Square, Kalantari Blvd., Mashhad 91331, Iran
E-mail: sahabnegahs@mums.ac.ir

*These authors contributed equally to this work.
Received June 15, 2023; Revised August 28, 2023; Accepted October 6, 2023.
This is an open-access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/), which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
Abstract
Despite enormous efforts, no effective medication has been found to significantly halt or even slow the progression of neurological diseases, such as acquired (e.g., traumatic brain injury, spinal cord injury, etc.) and chronic (e.g., Parkinson’s disease, Alzheimer’s disease, etc.) central nervous system disorders. So, researchers are looking for alternative therapeutic modalities to manage the disease’s symptoms and stop it from worsening. Concerning disease-modifying capabilities, stem cell therapy has emerged as an expanding domain. Among different types of stem cells, human endometrial regenerative cells have excellent regenerative properties, making them suitable for regenerative medicine. They have the potential for self-renewal and differentiation into three types of stem cells: epithelial stem cells, endothelial side population stem cells, and mesenchymal stem cells (MSCs). ERCs can be isolated from endometrial biopsy and menstrual blood samples. However, there is no comprehensive evidence on the effects of ERCs on neurological disorders. Hence, we initially explore the traits of these specific stem cells in this analysis, followed by an emphasis on their therapeutic potential in treating neurological disorders.
Keywords : Stem cell transplantation, Central nervous system diseases, Endometrium, Regenerative medicine
Introduction

One of the leading causes of disability and death globally is neurological disorders (1). Cerebrovascular disorders, Parkinson’s disease (PD), Alzheimer’s disease (AD), multiple sclerosis, motor neuron diseases (MNDs), and spinal cord injury (SCI) are common neurological diseases, which contribute the highest burden in disability (2). Current pharmacotherapy, for instance, at the first line for PD symptoms provides reasonable benefits at controlling the symptoms of the condition; however, the efficacy of the long-term use of drugs, despite increasing the dosage or doing brain surgical procedures, reduces with time (3-5). Hence, there is a need for further therapeutic advancements to reduce disease symptoms, slow or halt disease progression, and ultimately reverse the damaging effects on the nervous system.

Numerous studies have shown that stem cell therapy can be considered a potential novel treatment for neurodege-nerative diseases based on their self-renewal ability and differentiation capacity into different neural lineage cells (6, 7). Stem cells can reduce neurological disease progression rate and repair the chaotic microenvironment (8). These cells can continuously produce new cells and help repair organ damage depending on the organ features they reside in (8). They are categorized based on their potential differentiation ability into totipotent, pluripotent, multipotent, oligopotent, and unipotent (9). Stem cells can be isolated from embryonic and adult sources (10). Embryonic stem cells (ESCs) are pluripotent and are located in the inner cell layer of a pre-implanted embryo, which differentiate into all germinal layers (8). On the other hand, adult stem cells are found in various tissues like placenta, blood, bone marrow, skin, intestine, and brain (9, 11). Moreover, adult stem cells have the advantage of being sourced from the patient’s own body. This enables autologous transplantation, in which the patient receives stem cells that have been expanded or manipulated. Using the patient’s own cells reduces the risk of rejection or immunological complications, making autologous transplantation a more feasible treatment option. Additionally, adult stem cells carry a lower risk of tumor formation or uncontrolled growth than ESCs (12, 13). Therefore, prior to clinical practice, several features of stem cells must be pointed out, e.g., tumorigenicity, availability, immunogenicity, and differentiation capacity. Among different types of adult stem cells, human stem cells derived from the endometrium are highly regenerative sources of stem cells with ideal characteristics that make them an appropriate choice in regenerative medicine (14). They are also compatible with the International Society for Cellular Therapies (14).

Human endometrial regenerative cells (ERCs) consist of three types of stem cells: epithelial stem cells, endothelial side population (ESP) stem cells, and mesenchymal stem cells (MSCs) (15). ERCs can be obtained from human menstrual blood, known as menstrual blood-derived stem cells (MenSCs), and from the endometrium tissue, known as endometrium-derived stem cells (EnSCs), through biopsy. ERCs have the capacity to differentiate into various lineages, including adipocytes, osteocytes, cardiomyocytes, neurocytes, respiratory epithelial cells, endothelial cells, myocytes, hepatic cells, pancreatic cells, chondrocytes, fibroblasts, smooth muscle cells, neuron-like cells, glial-like cells, germ-like cells, various glandular lineages, and endometrial organoids (16-18). ERCs have been studied in various preclinical studies involving cardiac diseases, reproductive disorders, uterine abnormalities, diabetes, osteoporosis, and liver disease. Additionally, numerous clinical studies have been conducted on Duchene muscular dystrophy, heart failure, and multiple sclerosis (19, 20). The results of these studies are promising for the future of stem cell therapy (21-23).

In one study, intrathecal and intravenous administration of human EnSCs (hEnSCs) in four multiple sclerosis patients with 12 months of follow-up showed safety and efficacy in managing disease progression (19). Furthermore, hEnSCs have immunomodulatory effects by reducing inflammatory T-cells (19, 24). However, there has been limited attention given to the beneficial effects of ERCs in neurological diseases. Therefore, we have written this review to outline the main features of ERCs and highlight their therapeutic potential in addressing neurological disorders.

An Overview of Characteristics of ERCs

Before delving into a more detailed account of the ERCs, some general understanding of endometrium tissue is useful. The endometrium, which consists of the functional (the upper layer) and the basalis (adjacent to the myometrium), undergoes significant cell proliferation during tissue homeostasis and repair capabilities and remains intact during the menstrual period while generating new functional layers (18, 25). Stem cells in the human endometrium have been categorized into three types: epithelial stem/progenitor cells, ESP stem cells, and endometrial MSCs (18). However, a lack of exclusive gene or signaling pathway markers specific to endometrial tissue leads to significant overlap with other somatic stem cell types and even cancer stem cells. Nonetheless, endometrial MSCs can be classified into two categories based on expression pattern, i.e., CD140b/CD146 endometrial MSCs and sushi domain containing 2 (SUSD2) positive MSCs. These cells are located at the perivascular area. When dig deep on two MSCs, they are different in other markers, e.g., CD140b/CD146 endometrial MSCs are positive for CD29, CD44, CD73, CD90, and CD105, while negative for CD31, CD34, and CD45. On the other hand, SUSD2 positive-MSCs are positive for CD29, CD44, CD73, CD90, CD105, CD117, CD140b, CD146, STRO-1, and nucleoside triphosphate diphosphohydrola-se-2, but they don’t express CD31 and CD45 markers. However, it should be noted that some markers are shared between these two types of MSCs. Identifying specific markers and characteristics can help distinguish these cells for their intended applications. Nevertheless, ESP stem cells demonstrate the presence of CD90, CD105, CD73, CD45, CD34, CD31, CD133, STRO-1, and vimentin markers, while lacking CD13, CD105, estrogen receptor alpha, or progesterone receptor. Likewise, epithelial stem/progenitor cells exhibit the expression of N-cadherin, stage-specific embryonic antigen-1, and axin 2 (18, 26, 27).

Repetitive proliferation-menstruation cycles of uterus decline stem cells existed in the endometrium (26). It has been proved that MenSCs exist in deciduous endometrium, which might have a similar nature to EnSCs (27). One study showed the presence of endometrial clonogenic cells and SUSD2+ MSCs in females’ menstrual blood, even if they have endometriosis or not (28). These cells can be obtained with a simple uterine biopsy or by collecting human menstrual bleeding from menstrual cups. Therefore, the accessibility of these cells reduces the cost of cell obtaining (29) and can provide an unlimited source for more people with different socioeconomic levels. ERCs availability gives a favorable context for repetitive cell therapies (15, 30). Despite availability, these cells have an excellent potential to differentiate into multi-lineage sources (i.e., mesodermal, endodermal, and ectodermal lineages) (15, 29-32). They can, therefore, be a promising candidate for neuronal regenerative treatments compared to the other sources. Fig. 1 illustrates the various stem cells that can be isolated from the uterus and their characteristics regarding their potential for neural cell differentiation. Besides that, producing more stem cells at low passage (<8) is a central feature of self-renewal capacity that makes them the best candidate for a clinical trial. For MenSCs, a female can provide stem cells for 20,000 patients. Approximately 0.5×105 MenSCs can be derived from 5 ml menstrual blood. It has been shown that one hundred million MenSCs can be harvested at passage 3 (22).

Figure 1. Schematic diagram of isolation, characterization, and differentiation capacity of human endometrial regenerative cells (ERCs). Human ERCs can be collected from the uterus using an endometrial biopsy device and menstrual silicon cups. ERCs comprise three types of stem cells: epithelial stem cells, endothelial side population stem cells, and mesenchymal stem cells. ERCs have the ability to differentiate into various neural lineages. Created with BioRender.com.

However, MenSCs have a faster proliferation in early passages than late passages. Furthermore, cells derived from younger females have a more proliferative capacity (30). They could grow by a factor of two than bone marrow-derived MSCs (22). These cells become homogenous and make a spindle shape after 3 to 4 passages (32, 33). EnSCs have lower immunogenicity and tumor tropism than other MSCs (15).

Another significant aspect of cell therapy in neurological disorders is neural differentiation. Many complex pathways and molecules participate in neural trans-differentiation and specific neural cell type differentiation (34). Differentiation of hEnSCs into cholinergic neurons was made by nerve growth factor (NGF) and basic fibroblast growth factor (bFGF). It has been indicated that hEnSCs express receptors for NGF like neural stem cells (34). One study assessed oleic acid for improving neurogenesis capacity of hEnSCs (33). The mRNA levels of NF and β-TUBULIN as the neurogenesis markers in hEnSCs treated with oleic acid were significantly increased. Additionally, oleic acid increased the expression of ChAT and NF in hEnSCs, indicating hEnSCs have a great neuronal differentiation capacity (33). As shown in Table 1 (24, 35-43),

Table 1 . Human ERCs in the course of neurological models

SampleType of studyModelCell sourceRoute and no. of injectionsNo. of cellsResultMain findingReference
1By curettage from reproductive-aged females undergoing surgeryIn vitroMPTP mouse model of PDAdult hEnSCs--Exhibited neurogenic morphology, expression of the nestin and dopamine production TH and electrophysiological properties of neuronshEnSCs differentiated into the dopamine-producing neurons(35)
In vivoAdult hEnSCsStriatum 4 injections105 cellsExhibit nestin, and human TH, survive in the location they are transplanted and spontaneously migrate to areas of damage and spontaneously differentiate in vivo
2Collected by hysterectomy and curettage from eight monkeysIn vivoMPTP monkey model of ParkinsonMonkey EnSCsStriatum 2 injections into right striatum4×106 cells
Ability to migrate from the striatum and engraft in the substantia nigra. Also show significant migration to the contralateral sideEnSCs can migrate to the foci of cellular injury and differentiate to TH (+) neuron-like cells and protect endogenous dopaminergic neurons(40)
3Menstrual blood from healthy females donorsIn vitroOn MPP+ treated cellsMenSCs-CM--Significantly reduce ROS generation and significantly reduce the number of cells in late apoptosis stageMenSCs-CM can protect MPPC-induced cytotoxicity via reducing inflammation, oxidative stress, apoptosis and rescuing mitochondrial membrane potential(41)
4Collected with a Diva Cup from healthy femalesIn vivoAPP/PS1 transgenic mouse model of AlzheimerhMenSCsBilaterally into the hippocampus105 cellsReduced Ab deposition and decreased tau hyperphosphorylationDecreased tau hyperphosphorylation(36)
5Menstrual blood sampleIn vitroOGD injury model of strokeMenSCs--Cells were Nestin-positive, and readily differentiated into intermediate neuronal and astrocytic phenotype. Also, cells significantly reduced cell death and improved cell survival and elevated levels of trophic factors, such as VEGF, BDNF, and NT3The present neurostructural and behavioral benefits afforded by transplanted MenSCs(37)
In vivoRat model of strokeMenSCsIC (3 injections, striatum), IV4×105 for IC, 4×106 for IVsignificantly reduced behavioral abnormalities and increased survival of host cells
6Endometrial biopsyIn vivoMice model of autoimmune encephalomy-elitishEnSCsIntraperitoneally1×106Suppress neuroinflammationhEnSCs as a potent immunomodulatory tool for the treatment of autoimmune or neurodegenerative diseases(24)
7Uterine tubes and uterus were extracted from C57BL/6 WT and IDO−/−In vitroCo-culture of murine EnSCs and CD4+ T lymphocytesMurine EnSCs--Reduced overall inflammation in the CNS, including mononuclear cells infiltrate, cytokine secretion and microglial activationSuppressive activity of the unexplored murine EnSCs population, and shows the mechanism depends on IDO-Kynurenines-Aryl hydrocarbon receptor (AhR) axis(42)
In vivoMurine model of autoimm-une encephal-omyelitisMurine EnSCsIntraperitoneally1×106 cells on days 0 and 10 post-immuniza-tion
8Menstrual blood specimens were collected with menstrual cupsIn vivoRat model of hemisected SCIhMenSCsInjection into the injured site1×105Improved the locomotor function, reduced the inflammatorycell infiltrations and vacuolization in the lesion site, increased neuronal markers in the lesion area, enhanced expression and secretion of BDNF, reduced scar formation, and decreased the expression of inflammatory cytokinesMenSCs transplantation promotion of the functional recovery of SCI rats via enhanced expression and secretion of BDNF, reduced scar formation and decreased the expression of inflammatory cytokine(38)
9Human endometrial biopsy
In vivoMouse model of hippocampal injuryEnSCs-derived extracellular vesiclesIntranasalTotal of 500 mg of EVs (released by 5×106 eMSCs) protein per kilogram of animal weightPrevented histological damage and preserved speed locomotion and displacement changes presumably due to the growth factors contained in those vesiclesIntranasal administration of EnSCs – EVs could improve recovery of hippocampal tissue(43)
10Menstrual bloodIn vivoMouse SCI modelhMenSCsIntrathecal1×105 cell/μl and 2×105 cellMenSCs transplantation Shh–induced MenSCs accelerated neuronal recovery, inhibited the formation ofglial cells and microglial activation atthe injured site, inhibited the expression of inflammatory factors, and improved the inflammatory microenvironment to achieve functional recovery of SCIMenSCs transplantation improved functional recovery(39)

The main characteristics of experimental research in the application of endometrial regenerative cells (ERCs) in some neurological models.

PD: Parkinson’s disease, hEnSCs: human endometrial derived stem cells, TH: tyrosine hydroxylase, MenSCs: menstrual blood-derived stem cells, MenSCs-CM: conditioned medium of human menstrual blood derived endometrial stem cells, ROS: reactive oxygen species, OGD: oxygen glucose deprivation, VEGF: vascular endothelial growth factor, BDNF: brain-derived neurotrophic factor, NT3: neurotrophin-3, SCI: spinal cord injury, EVs: extracellular vesicles.



hEnSCs differentiation into dopamine-producing neurons is described. It is proved that hEnSCs are promising cell sources for neural tissue engineering (44).

ERCs in the Context of Neurological Disorders

Based on our research in the field of ERCs, we conducted a comprehensive review to examine the effects of these types of stem cells on various neurological disorders. Specifically, we focused on conditions such as PD, AD, MND, as well as acquired brain injuries such as stroke and traumatic brain injury (TBI), by analyzing published articles related to these conditions.

The most common neurodegenerative movement disorder is PD, characterized by the depletion of dopaminergic neurons and the aggregation of misfolded alpha-synuclein, leading to motor and non-motor symptoms (3). Administration of hEnSCs in PD animal models has significantly increased dopamine concentration (35). In vitro studies have also demonstrated the ability of hEnSCs to differentiate into dopaminergic neurons that exhibit morphological and molecular characteristics, such as pyramidal cell bodies, axon and dendrite projections, and synapse formation. These cells express nestin and tyrosine hydroxylase, which are neural markers and critical enzymes in dopamine synthesis. Furthermore, hEnSCs show a reasonable survival rate after transplantation and exhibit migration capacity to the damaged region (35).

Several categories can be identified when examining the mechanisms of action for ERCs in PD. These stem cells have shown the ability to inhibit inflammatory pathways, reducing the expression of inflammatory markers, such as COX-2, interleukin (IL)-1β, IL-6, inducible nitric oxide synthase (iNOS), and tumor necrosis factor α (TNFα). Additionally, MenSCs secrete neurotrophic agents such as NGF, brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT3), and NT4/5, which can reduce apoptosis in PD models. MenSCs also express members of the GDNF family of ligands, including ARTN, GDNF, NTN, and PSPN, as well as other dopaminergic neurotrophic factors that enhance the survival of dopaminergic neurons. Furthermore, MenSCs can restore mitochondrial membrane potential, which is often disrupted in PD pathogenesis (3, 27).

In AD, the main pathology is believed to be an imbalance between oxidant and antioxidant processes because of the deposition of amyloid-beta peptides and the activation of microglia (36). This imbalance leads to progressive neuronal loss, resulting in the clinical manifestations of AD. Administering MenSCs has been shown to convert microglia activity to a non-inflammatory state by reducing pro-inflammatory cytokines like IL-1β and TNFα, while increasing anti-inflammatory markers such as IL-4, YM1, Arg1, Fizz1, and CD206 in the hippocampus and cortex of AD models. This conversion of microglia leads to the production of insulin-degrading enzyme and neprilysin, which degrades amyloid-beta plaques. Another mechanism involves the inhibition of the β-secretase enzyme (36). MenSCs also inhibit glycogen synthase kinase-3b after transplantation, reducing tau hyperphosphorylation, a characteristic feature of AD (36). Amyotrophic lateral sclerosis (ALS), the most common neurodegenerative disease in the middle ages, is classified as a MND characterized by the degeneration of motor neurons in the central nervous system. Unfortunately, this condition leads to death within 3 to 5 years (45). While there is currently no systematic study explicitly exploring the effects of hEnSCs and MenSCs on ALS models, a review of the existing literature suggests that hEnSCs may hold promise as a potential treatment for ALS.

For instance, a comparative study demonstrated that hEnSCs exhibited a higher capacity for differentiation into motor neurons than human bone marrow stem cells (46). Additionally, in vitro studies have shown that hEnSCs express several motor neuron markers, such as ISL‐1, CHAT, β-tubulin III, NF-H, HB9, pax6, Islet-1, TUJ-1, NES, NEFH, and SYP (46-49). In ALS, it is imperative to utilize stem cells that can form synaptic connections with muscle cells. In an experimental study, hEnSCs grafted inside a conduit made of poly ε-caprolactone (PCL)/Collagen/nanobioglass (NBG) not only demonstrated successful grow-th and connection with damaged nerve regions but also exhibited improved synapse formation with muscle cells (50).

To fully understand the potential of ERCs in ALS models, further investigation is needed to evaluate their effects on maintaining axonal integrity, besides their neuropro-tective properties and ability to differentiate into motor neurons. Table 2 summarizes the potential differentiation of hEnSCs and MenSCs into motor neurons. The last category that we reviewed was acquired brain injuries including stroke and TBI, which have high mortality rates and long-lasting disability. The pathological changes are categorized into primary and secondary injuries. Solid evidence demonstrates that a variety of cellular and molecular alterations play vital roles in connecting primary injury to the secondary injury, e.g., inflammatory pathways

Table 2 . The differentiation capacity of hEnSCs into motor neuron

Cell typeScaffoldCell numberMarkersDuration of differentiation (day)% Cell expression markerMechanismBeneficial resultMain findingReference
hEnSCsTissue culture polystyrene (TCP) and poly ε-caprolactone (PCL) nanofibrous scaffold5×104 cellsISL‐1, CHAT, islet-1, Pax6, NF-H, Hb9, and β‐tubulin III1540∼90Significant upregulation of motor neuron markersExhibiting neural morphology and interconnection of cellsImproved motor neuron differentiation(46)
hEnSCsBiodegradable PLGA nanofiber scaffolds5×104 cellsISL‐1, CHAT, β-tubulin III
NF-H
HB9, pax6
1540∼89Increased gene expression of markersExhibiting neural morphology and interconnection of cellsImproved motor neuron differentiation(48)
hEnSCsElectro spun Biocomposite Polycaprolactone/Collagen scaffolds5×104 cellsNF-H, TUJ-1, Chat, Islet-1, and HB91560∼80Inhibition of the PI3K/Akt pathway
Neural marker expression
Exhibiting neural morphology characteristicsImproved motor neuron differentiation(47)
hEnSCsEpothilone B (EpoB)2×105 cellsISL‐1, CHAT, and HB9
NES
NEFH
SYP, and β‐tubulin III proteins
15More than 90Upregulation of motor neuron markersImproved axonal length and neuronal alignmentImproved motor neuron differentiation(49)

Differentiation capabilities into motor neuron is a promising function of human endometrium-derived stem cells (hEnSCs) for the treatment of motor related disorders.



and oxidative stress are the key players in expanding the pathological changes in relation to secondary injury. Moreover, the presence of cavity lesions resulting from mechanical trauma, such as TBI and SCI, poses a significant obstacle to treatment. Regarding this concept, stem cells with high bystander effects can be a best therapeutic option. Particularly, promising results were observed after transplantation of MenSCs by secreting neurotrophic factors, such as vascular endothelial growth factor (VEGF), BDNF, and NT3 (37). Furthermore, promising findings have been observed in the reduction of cavity lesions using MenSCs in a spinal cord hemisection model. Cellular assessments revealed increased axonal regeneration and decreased expression of chondroitin sulfate proteoglycans in the MenSCs group compared to the control group. Additionally, MenSCs transplantation was found to inhibit the expression of TNFα and IL-1β (38). Notably, in an SCI model, the rate of motor function recovery was found to correlate with the differentiation of oligodendrocyte progenitor cells in the MenSCs group (51). Furthermore, MenSCs transplantation enhanced motor function recovery in SCI animals and stimulated the immune response and defense. Transcriptome sequencing and analysis revealed the close association of SCI with immu-ne system processes. The transplantation of MenSCs was closely linked to pathways involved in the differentiation of Th1, Th2, and Th17 cells. Additionally, MenSCs regulated the differentiation and activation of Th2 and M2 macrophages, produced anti-inflammatory factors, and attenuated the inflammatory response at the injury site, thereby promoting SCI function recovery. In the MenSCs transplantation group, the expression of iNOS and CD206 in microglia decreased, indicating reduced inflammation. Moreover, MenSCs improved the balance between M1 and M2 microglia within the highly inflammatory microenvironment (39).

Additionally, positive outcomes can be achieved when combining hEnSCs with different scaffolds. In the case of differentiating hEnSCs seeded on PCL/gelatin scaffolds, enhanced neuronal regeneration, increased motor function, and axonal remyelination were observed in an SCI model (52). Furthermore, hEnSCs seeded on PCL demonstrated a reduction in the secondary response to injury and an improvement in motor function (53). The potential therapeutic effects of hEnSCs and MenSCs in acquired brain injuries are summarized in Fig. 2. Considering these findings, these cells can be considered suitable candidates for stem cell therapy in the context of acquired brain injuries.

Figure 2. Mechanisms of probable therapeutic effects of human endometrial regenerative cells (hERCs) in a traumatic brain injury model. hERCs can alleviate detrimental effects of brain injury through several important pathways; 1. Increasing the expression of neurotrophic factors, such as nerve growth factor, brain-derived neurotrophic factor, and neurotrophin-3 for triggering the neurogenesis; 2. Improving the volume of lesion cavity; 3. Decreasing the expression levels of proinflammatory cytokines in the brain tissue and prevention of neuroinflammation; 4. Reducing the expression of proapoptotic genes, such as Bad and Bax; 5. Increasing nerve regeneration and improving the axonal remyelination; 6. Decreasing the leukocytes infiltration following inflammatory condition; 7. Suppressing the main inflammatory cytokines and reactive oxygen species (ROS); 8. Restoring the reactive glial cells and controlling vasogenic and cytotoxic edema with reduction of proinflammatory cytokines and ROS; 9. Inducing the formation of micro vessels. BBB: blood-brain barrier, IL-1β: interleukin 1 beta, IL-6: interleukin 6, TNFα: tumor necrosis factor α, COX2: cyclooxygenase 2, iNOS: inducible nitric oxide synthase.
Conclusion

Accessibility and high survival rates after transplantation are key characteristics for the clinical use of stem cells. Thus, the search for readily available stem cells with excellent survival and differentiation capabilities becomes crucial in translating stem cell applications to benefit patients. In this context, we investigated the potential of ERCs as promising candidates for treating neurological disorders. We reviewed the current evidence and discussed the beneficial effects of ERCs in this area. ERCs have shown the ability to alleviate pathological changes by inhibiting inflammatory and apoptotic pathways. Moreover, they secrete various neuroprotective agents that promote remyelination, axonal growth, and improve synaptic integrity and extracellular remodeling. Considering the available data, it is essential for future research to carefully examine the potential effects of ERCs, including evaluating their safety and efficacy at the clinical level.

Acknowledgments

We would like to thank the Clinical Research Development Unit, Imam Reza Hospital, Mashhad University of Medical Sciences, for their assistance in this manuscript.

Potential Conflict of Interest

There is no potential conflict of interest to declare.

Data Availability Statement

All data analyzed during this study are included in this published article.

Authors’ Contribution

Conceptualization: SSN. Data curation: SSN, JM. Investigation: JM, EN, RAA. Methodology: SSN, JM. Project administration: SSN. Resources: JM. Software: AS. Supervision: SSN. Validation: SSN. Visualization: AS. Writing – original draft: JM, EN, SSN. Writing – review and editing: SSN, JM.

Funding

None.

References
  1. Feigin VL, Vos T, Nichols E, et al. The global burden of neurological disorders: translating evidence into policy. Lancet Neurol 2020;19:255-265.
    Pubmed KoreaMed CrossRef
  2. De Gioia R, Biella F, Citterio G, et al. Neural stem cell transplantation for neurodegenerative diseases. Int J Mol Sci 2020;21:3103.
    Pubmed KoreaMed CrossRef
  3. Balestrino R, Schapira AHV. Parkinson disease. Eur J Neurol 2020;27:27-42.
    Pubmed CrossRef
  4. Chiken S, Nambu A. Mechanism of deep brain stimulation: inhibition, excitation, or disruption? Neuroscientist 2016;22:313-322.
    Pubmed KoreaMed CrossRef
  5. Fox SH, Katzenschlager R, Lim SY, et al. The Movement Disorder Society evidence-based medicine review update: treatments for the motor symptoms of Parkinson’s disease. Mov Disord 2011;26 Suppl 3:S2-S41.
  6. Ahmadian-Moghadam H, Sadat-Shirazi MS, Zarrindast MR. Therapeutic potential of stem cells for treatment of neurodegenerative diseases. Biotechnol Lett 2020;42:1073-1101.
    Pubmed CrossRef
  7. Andrzejewska A, Dabrowska S, Lukomska B, Janowski M. Mesenchymal stem cells for neurological disorders. Adv Sci (Weinh). 2021;8:2002944.
    Pubmed KoreaMed CrossRef
  8. Zakrzewski W, Dobrzyński M, Szymonowicz M, Rybak Z. Stem cells: past, present, and future. Stem Cell Res Ther 2019;10:68.
    Pubmed KoreaMed CrossRef
  9. Kolios G, Moodley Y. Introduction to stem cells and regenerative medicine. Respiration 2013;85:3-10.
    Pubmed CrossRef
  10. Sobhani A, Khanlarkhani N, Baazm M, et al. Multipotent stem cell and current application. Acta Med Iran 2017;55:6-23.
    Pubmed
  11. Gurusamy N, Alsayari A, Rajasingh S, Rajasingh J. Adult stem cells for regenerative therapy. Prog Mol Biol Transl Sci 2018;160:1-22.
    Pubmed CrossRef
  12. Barkho BZ, Zhao X. Adult neural stem cells: response to stroke injury and potential for therapeutic applications. Curr Stem Cell Res Ther 2011;6:327-338.
    Pubmed KoreaMed CrossRef
  13. Nam H, Lee KH, Nam DH, Joo KM. Adult human neural stem cell therapeutics: current developmental status and prospect. World J Stem Cells 2015;7:126-136.
    Pubmed KoreaMed CrossRef
  14. Bozorgmehr M, Gurung S, Darzi S, et al. Endometrial and menstrual blood mesenchymal stem/stromal cells: biologi-cal properties and clinical application. Front Cell Dev Biol 2020;8:497.
    Pubmed KoreaMed CrossRef
  15. Zuo W, Xie B, Li C, et al. The clinical applications of endometrial mesenchymal stem cells. Biopreserv Biobank 2018;16:158-164.
    Pubmed KoreaMed CrossRef
  16. Meng X, Ichim TE, Zhong J, et al. Endometrial regenera-tive cells: a novel stem cell population. J Transl Med 2007;5:57.
    Pubmed KoreaMed CrossRef
  17. Jin W, Zhao Y, Hu Y, et al. Stromal cell-derived factor-1 enhances the therapeutic effects of human endometrial regenerative cells in a mouse sepsis model. Stem Cells Int. 2020;2020:4820543.
    Pubmed KoreaMed CrossRef
  18. Kong Y, Shao Y, Ren C, Yang G. Endometrial stem/progenitor cells and their roles in immunity, clinical application, and endometriosis. Stem Cell Res Ther 2021;12:474.
    Pubmed KoreaMed CrossRef
  19. Zhong Z, Patel AN, Ichim TE, et al. Feasibility investi-gation of allogeneic endometrial regenerative cells. J Transl Med 2009;7:15.
    Pubmed KoreaMed CrossRef
  20. Ichim TE, Alexandrescu DT, Solano F, et al. Mesenchymal stem cells as anti-inflammatories: implications for treatment of Duchenne muscular dystrophy. Cell Immunol 2010;260:75-82.
    Pubmed CrossRef
  21. Wang Z, Wang D, Liu Y, et al. Mesenchymal stem cell in mice uterine and its therapeutic effect on osteoporosis. Rejuvenation Res 2021;24:139-150.
    Pubmed CrossRef
  22. Liu Y, Niu R, Li W, et al. Therapeutic potential of mens-trual blood-derived endometrial stem cells in cardiac diseases. Cell Mol Life Sci 2019;76:1681-1695.
    Pubmed KoreaMed CrossRef
  23. Chen L, Qu J, Xiang C. The multi-functional roles of menstrual blood-derived stem cells in regenerative medicine. Stem Cell Res Ther 2019;10:1.
    Pubmed KoreaMed CrossRef
  24. Peron JP, Jazedje T, Brandão WN, et al. Human endometrial-derived mesenchymal stem cells suppress inflam-mation in the central nervous system of EAE mice. Stem Cell Rev Rep 2012;8:940-952.
    Pubmed CrossRef
  25. Hong IS. Endometrial stem/progenitor cells: properties, origins, and functions. Genes Dis 2022;10:931-947.
    Pubmed KoreaMed CrossRef
  26. Maruyama T. Endometrial stem/progenitor cells. J Obstet Gynaecol Res 2014;40:2015-2022.
    Pubmed CrossRef
  27. Li H, Yahaya BH, Ng WH, Yusoff NM, Lin J. Conditioned medium of human menstrual blood-derived endometrial stem cells protects against MPP-induced cytotoxicity in vitro. Front Mol Neurosci 2019;12:80.
  28. Masuda H, Schwab KE, Filby CE, et al. Endometrial stem/progenitor cells in menstrual blood and peritoneal fluid of women with and without endometriosis. Reprod Biomed Online 2021;43:3-13.
    Pubmed CrossRef
  29. Mobarakeh ZT, Ai J, Yazdani F, et al. Human endometrial stem cells as a new source for programming to neural cells. Cell Biol Int Rep (2010). 2012;19:e00015.
    Pubmed KoreaMed CrossRef
  30. Liu Y, Niu R, Yang F, et al. Biological characteristics of human menstrual blood-derived endometrial stem cells. J Cell Mol Med 2018;22:1627-1639.
    Pubmed KoreaMed CrossRef
  31. Cheng Y, Li L, Wang D, et al. Characteristics of human endometrium-derived mesenchymal stem cells and their tropism to endometriosis. Stem Cells Int. 2017;2017:4794827.
    Pubmed KoreaMed CrossRef
  32. Fayazi M, Salehnia M, Ziaei S. Differentiation of human CD146-positive endometrial stem cells to adipogenic-, osteogenic-, neural progenitor-, and glial-like cells. In Vitro Cell Dev Biol Anim 2015;51:408-414.
    Pubmed CrossRef
  33. Kojour MA, Ebrahimi-Barough S, Kouchesfehani HM, Jalali H, Ebrahim MH. Oleic acid promotes the expression of neural markers in differentiated human endometrial stem cells. J Chem Neuroanat 2017;79:51-57.
    Pubmed CrossRef
  34. Noureddini M, Verdi J, Mortazavi-Tabatabaei SA, et al. Human endometrial stem cell neurogenesis in response to NGF and bFGF. Cell Biol Int 2012;36:961-966.
    Pubmed CrossRef
  35. Wolff EF, Gao XB, Yao KV, et al. Endometrial stem cell transplantation restores dopamine production in a Parki-nson’s disease model. J Cell Mol Med 2011;15:747-755.
  36. Zhao Y, Chen X, Wu Y, Wang Y, Li Y, Xiang C. Trans-plantation of human menstrual blood-derived mesenchymal stem cells alleviates Alzheimer’s disease-like pathology in APP/PS1 transgenic mice. Front Mol Neurosci 2018;11:140.
  37. Borlongan CV, Kaneko Y, Maki M, et al. Menstrual blood cells display stem cell-like phenotypic markers and exert neuroprotection following transplantation in experimental stroke. Stem Cells Dev 2010;19:439-452.
    Pubmed KoreaMed CrossRef
  38. Wu Q, Wang Q, Li Z, et al. Human menstrual blood-derived stem cells promote functional recovery in a rat spinal cord hemisection model. Cell Death Dis 2018;9:882.
    Pubmed KoreaMed CrossRef
  39. Shi Y, Liu Y, Zhang B, Li X, Lin J, Yang C. Human menstrual blood-derived endometrial stem cells promote functional recovery by improving the inflammatory microenvi-ronment in a mouse spinal cord injury model. Cell Trans-plant. 2023;32:9636897231154579.
    Pubmed KoreaMed CrossRef
  40. Wolff EF, Mutlu L, Massasa EE, Elsworth JD, Eugene Redmond D Jr, Taylor HS. Endometrial stem cell transplantation in MPTP- exposed primates: an alternative cell source for treatment of Parkinson’s disease. J Cell Mol Med 2015;19:249-256.
  41. Yang X, Devianti M, Yang YH, et al. Endometrial mesenchymal stem/stromal cell modulation of T cell prolifera-tion. Reproduction 2019;157:43-52.
    Pubmed CrossRef
  42. Manganeli Polonio C, Longo de Freitas C, Garcia de Oliveira M, et al. Murine endometrial-derived mesenchymal stem cells suppress experimental autoimmune encephalomyelitis depending on indoleamine-2,3-dioxygenase expression. Clin Sci (Lond) 2021;135:1065-1082.
    Pubmed CrossRef
  43. León-Moreno LC, Castañeda-Arellano R, Aguilar-García IG, et al. Kinematic changes in a mouse model of penetrating hippocampal injury and their recovery after intranasal administration of endometrial mesenchymal stem cell-derived extracellular vesicles. Front Cell Neurosci. 2020;14:579162.
    Pubmed KoreaMed CrossRef
  44. Hasanzadeh E, Ebrahimi-Barough S, Mahmoodi N, et al. Defining the role of 17β-estradiol in human endometrial stem cells differentiation into neuron-like cells. Cell Biol Int 2021;45:140-153.
    Pubmed CrossRef
  45. Brown RH, Al-Chalabi A. Amyotrophic lateral sclerosis. N Engl J Med 2017;377:162-172.
    Pubmed CrossRef
  46. Shirian S, Ebrahimi-Barough S, Saberi H, et al. Compari-son of capability of human bone marrow mesenchymal stem cells and endometrial stem cells to differentiate into motor neurons on electrospun poly (ε-caprolactone) scaffold. Mol Neurobiol 2016;53:5278-5287.
    Pubmed CrossRef
  47. Ebrahimi-Barough S, Hoveizi E, Yazdankhah M, et al. Inhibitor of PI3K/Akt signaling pathway small molecule promotes motor neuron differentiation of human endome-trial stem cells cultured on electrospun biocomposite polycaprolactone/collagen scaffolds. Mol Neurobiol 2017;54:2547-2554.
    Pubmed CrossRef
  48. Ebrahimi-Barough S, Norouzi Javidan A, Saberi H, et al. Evaluation of motor neuron-like cell differentiation of hEnSCs on biodegradable PLGA nanofiber scaffolds. Mol Neurobiol 2015;52:1704-1713.
    Pubmed CrossRef
  49. Mahmoodi N, Ai J, Ebrahimi-Barough S, et al. Microtubule stabilizer epothilone B as a motor neuron differentiation agent for human endometrial stem cells. Cell Biol Int 2020;44:1168-1183.
    Pubmed CrossRef
  50. Mohamadi F, Ebrahimi-Barough S, Nourani MR, et al. Enhanced sciatic nerve regeneration by human endometrial stem cells in an electrospun poly (ε-caprolactone)/collagen/NBG nerve conduit in rat. Artif Cells Nanomed Biotechnol 2018;46:1731-1743.
    Pubmed CrossRef
  51. Jalali Monfared M, Nasirinezhad F, Ebrahimi-Barough S, et al. Transplantation of miR-219 overexpressed human endometrial stem cells encapsulated in fibrin hydrogel in spinal cord injury. J Cell Physiol 2019;234:18887-18896.
    Pubmed CrossRef
  52. Babaloo H, Ebrahimi-Barough S, Derakhshan MA, et al. PCL/gelatin nanofibrous scaffolds with human endometrial stem cells/Schwann cells facilitate axon regeneration in spinal cord injury. J Cell Physiol 2019;234:11060-11069.
    Pubmed CrossRef
  53. Terraf P, Kouhsari SM, Ai J, Babaloo H. Tissue-engineered regeneration of hemisected spinal cord using human endometrial stem cells, poly ε-caprolactone scaffolds, and crocin as a neuroprotective agent. Mol Neurobiol 2017;54:5657-5667.
    Pubmed CrossRef


November 2024, 17 (4)
Full Text(PDF) Free

Social Network Service

Services

Cited By Articles
CrossRef (0)